Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters











Publication year range
1.
Calcif Tissue Int ; 115(4): 432-444, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39030433

ABSTRACT

Renin-angiotensin-aldosterone system plays a crucial role in the regulation of blood pressure and fluid homeostasis. It is reported to be involved in mediating osteoclastogenesis and bone loss in diseases of inflammatory bone resorption such as osteoporosis. Angiotensin-(1-7), a product of Angiotensin I and II (Ang I, II), is cleaved by Angiotensin-converting enzyme 2 and then binds to Mas receptor to counteract inflammatory effects produced by Ang II. However, the mechanism by which Ang-(1-7) reduces bone resorption remains unclear. Therefore, we aim to elucidate the effects of Ang-(1-7) on lipopolysaccharide (LPS)-induced osteoclastogenesis. In vivo, mice were supracalvarial injected with Ang-(1-7) or LPS ± Ang-(1-7) subcutaneously. Bone resorption and osteoclast formation were compared using micro-computed tomography, tartrate-resistant acid phosphatase (TRAP) stain, and real-time PCR. We found that Ang-(1-7) attenuated tumor necrosis factor (TNF)-α, TRAP, and Cathepsin K expression from calvaria and decreased osteoclast number along with bone resorption at the suture mesenchyme. In vitro, RANKL/TNF-α ± Ang-(1-7) was added to cultures of bone marrow-derived macrophages (BMMs) and osteoclast formation was measured via TRAP staining. The effect of Ang-(1-7) on LPS-induced osteoblasts RANKL expression and peritoneal macrophages TNF-α expression was also investigated. The effect of Ang-(1-7) on the MAPK and NF-κB pathway was studied by Western blotting. As a result, Ang-(1-7) reduced LPS-stimulated macrophages TNF-α expression and inhibited the MAPK and NF-κB pathway activation. However, Ang-(1-7) did not affect osteoclastogenesis induced by RANKL/TNF-α nor reduce osteoblasts RANKL expression in vitro. In conclusion, Ang-(1-7) alleviated LPS-induced osteoclastogenesis and bone resorption in vivo via inhibiting TNF-α expression in macrophages.


Subject(s)
Angiotensin I , Bone Resorption , Macrophages , Mice, Inbred C57BL , Osteoclasts , Peptide Fragments , Tumor Necrosis Factor-alpha , Animals , Angiotensin I/pharmacology , Angiotensin I/metabolism , Bone Resorption/metabolism , Bone Resorption/prevention & control , Tumor Necrosis Factor-alpha/metabolism , Peptide Fragments/pharmacology , Osteoclasts/drug effects , Osteoclasts/metabolism , Mice , Macrophages/drug effects , Macrophages/metabolism , Male , Osteogenesis/drug effects , Lipopolysaccharides/pharmacology , Inflammation/metabolism
2.
Free Radic Biol Med ; 214: 69-79, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38336100

ABSTRACT

Cyclooxygenase-2 (COX-2) is an inducible enzyme responsible for prostaglandin synthesis during inflammation and immune responses. Our previous results show that NAD+ level decreased in activated macrophages while nicotinamide mononucleotide (NMN) supplementation suppressed the inflammatory responses via restoring NAD+ level and downregulating COX-2. However, whether NMN downregulates COX-2 in mouse model of inflammation, and its underlying mechanism needs to be further explored. In the present study, we established LPS- and alum-induced inflammation model and demonstrated that NMN suppressed the inflammatory responses in vivo. Quantitative proteomics in mouse peritoneal macrophages identified that NMN activated AhR signaling pathway in activated macrophages. Furthermore, we revealed that NMN supplementation led to IDO1 activation and kynurenine accumulation, which caused AhR nuclear translocation and activation. On the other hand, AhR or IDO1 knockout abolished the effects of NMN on suppressing COX-2 expression and inflammatory responses in macrophages. In summary, our results demonstrated that NMN suppresses inflammatory responses by activating IDO-kynurenine-AhR pathway, and suggested that administration of NMN in early-stage immuno-activation may cause an adverse health effect.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Kynurenine , Animals , Mice , Cyclooxygenase 2/genetics , Nicotinamide Mononucleotide , NAD , Macrophages , Inflammation , Signal Transduction , Dietary Supplements
3.
Microb Pathog ; 183: 106294, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37567327

ABSTRACT

The CXCL8/CXCR1 axis in conjoint with the free radicals and anti-oxidants dictates the severity of inflammation caused by the bacteria, Staphylococcus aureus. S.aureus mediated inflammatory processes is regulated by NF-κB and its product, iNOS. The objective of this study was to examine the effects of inhibition of NF-κB and iNOS on CXCL8/CXCR1, alteration in M1/M2 polarization of macrophages and associated inflammatory responses during S.aureus infection in vitro. For this, the murine peritoneal macrophages were pretreated with NF-κB inhibitor, Pyrrolidine dithiocarbamate (PDTC) and iNOS inhibitor, L-N-monomethyl arginine (LNMMA), either alone or in combination, followed by time-dependent S.aureus infection. The chemotactic migrations of macrophages were determined by the agarose spot assay. The iNOS, NF-κB and CXCR1 protein expressions were evaluated. The ROS level (superoxide, H2O2, NO) and antioxidant activities (SOD, CAT, GSH, arginase) were measured. The intra-macrophage phagoctyic activity had been analyzed by confocal microscopy. S.aureus activated macrophages showed increased iNOS expression that symbolizes M1 characterization of macrophages. The results suggest that the combination treatment of LNMMA + PDTC was effective in diminution of CXCL8 production and CXCR1 expression through downregulation of NF-κB and iNOS signaling pathway. Consequently, there was decrement in macrophage migration, reduced ROS generation, elevated antioxidant enzyme activity as well as bacterial phagocytosis at 90 min post bacterial infection. The increased arginase activity further proves the switch from pro-inflammatory M1 to anti-inflammatory M2 polarization of macrophages. Concludingly, the combination of PDTC + LNMMA could resolve S.aureus mediated inflammation through mitigation of CXCL8/CXCR1 pathway switching from M1 to M2 polarization.


Subject(s)
Macrophages, Peritoneal , Staphylococcal Infections , Mice , Animals , Macrophages, Peritoneal/microbiology , Staphylococcus aureus/metabolism , omega-N-Methylarginine/metabolism , Antioxidants/pharmacology , Antioxidants/metabolism , Reactive Oxygen Species/metabolism , NF-kappa B/metabolism , Hydrogen Peroxide/metabolism , Arginase/metabolism , Cytokines/metabolism , Staphylococcal Infections/microbiology , Receptors, Interleukin-8A/metabolism , Inflammation/metabolism
4.
Int Immunopharmacol ; 120: 110297, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37207443

ABSTRACT

Overexpression of Staphylococcus aureus mediated CXCL8/CXCR1 axis is a major cause of sepsis and severe inflammatory diseases. This chemokine acts conjointly with various pro-inflammatory and anti-inflammatory cytokines that govern the severity of inflammation. The effects of different combinations of exogenous cytokines on CXCR1 expression in macrophages remain undetermined. Exogenous cytokine and anti-inflammatory cytokine therapy had been used to modulate CXCL8 and CXCR1 expression in peritoneal macrophages. Male Swiss albino mice were inoculated with live S. aureus (106 cells/ mouse) for the development of infection. Exogenous cytokines (TNF-α, IL-12, IFN-γ and IL-10) were administered intraperitoneally (single or combination) 24 h post S. aureus infection. The mice were sacrificed and peritoneal macrophages were isolated three days post infection. CXCL8, IL-12, IL-10 secretion, ROS generation and the bacterial phagocytic process had been evaluated. Western blot was used to study the expressions of TNFR1, IL-1R, CXCR1 and NF-κB. TNF-α, IL-12 and IFN-γ treatments aggravated CXCL8 and CXCR1 expression in the macrophages of infected mice. TNF-α + IFN-γ treatment was a major inducer of nitric oxide release and mediated maximum bacterial killing. IL-12 + TNF-α treatment was most potent in increasing ROS, CXCL8/CXCR1 expression through increased levels of TNFR1, IL-1R and NF-κB activation. IL-10 reversed the effects of exogenous cytokines but also impaired the bacterial clearance phenomenon in peritoneal lavage. Treatment with IL-12 + TNF-α + IL-10 was most effective in ameliorating oxidative stress, reduced CXCL8 release and expression levels of TNFR1, IL-1R, and NF-κB. Concludingly, IL-12 + TNF-α + IL-10 treatment mitigated CXCL8/CXCR1 expression and inflammatory signalling via downregulation of TNFR1-IL-1R-NF-κB pathway in peritoneal macrophages and inflammatory sequelae during S. aureus infection.


Subject(s)
Receptors, Tumor Necrosis Factor, Type I , Staphylococcal Infections , Animals , Male , Mice , Cytokines/metabolism , Interleukin-10 , Interleukin-12/therapeutic use , Macrophages, Peritoneal/metabolism , NF-kappa B , Reactive Oxygen Species , Receptors, Interleukin-8A/metabolism , Receptors, Tumor Necrosis Factor, Type I/therapeutic use , Receptors, Tumor Necrosis Factor, Type I/metabolism , Staphylococcal Infections/metabolism , Staphylococcus aureus/metabolism , Tumor Necrosis Factor-alpha
5.
Methods Mol Biol ; 2562: 259-270, 2023.
Article in English | MEDLINE | ID: mdl-36272082

ABSTRACT

Salamanders have been used as research models for centuries. While they exhibit a wide range of biological features not seen in mammals, none has captivated scientists like their ability to regenerate. Interestingly, axolotl macrophages have emerged as an essential cell population for tissue regeneration. Whether the same is true in other salamanders such as newt species Notophthalmus viridescens, Cynops pyrrhogaster, or Pleurodeles waltl remains to be seen. Unfortunately, regardless of the species, molecular tools to study macrophage function in salamanders are lacking. We propose that the readily available, terminally differentiated peritoneal macrophages from newts or axolotls could be used to validate molecular reagents in the study of macrophage function during tissue regeneration in salamanders.


Subject(s)
Macrophages, Peritoneal , Urodela , Animals , Pleurodeles , Mammals , Salamandridae
6.
Front Immunol ; 13: 969468, 2022.
Article in English | MEDLINE | ID: mdl-36119051

ABSTRACT

Background: The peritoneal cavity contains many site-specific immune cells which constitute a unique immune microenvironment. However, it is unclear how the local immune signature is altered in patients with peritoneal metastases (PM). Methods: Peritoneal lavage fluid or ascites were obtained from 122 patients with various stages of gastric cancer (GC). Cells recovered from peritoneal fluids were immunostained with mAbs for lymphocyte-, macrophage- and tumor cell-specific antigens and the frequencies of leukocyte subsets and antigen expression levels were evaluated with multi-color flowcytometry. Results: The proportions of CD8(+) T cells, CD3(+)CD56(+) NKT-like cells, and CD3(-)CD56(+) NK cells to CD45(+) leukocytes were significantly reduced in patients with PM compared to those without PM. In patients with PM, the rates of CD8 (+) T cells and NKT-like cells correlated inversely with the tumor leukocyte ratio (TLR), the relative frequency of CD326(+) tumor cells to CD45(+) leukocytes. In contrast, the proportion of CD19(+) B cells was significantly increased in patients with PM, and their proportion correlated positively with the TLR and peritoneal carcinomatosis index (PCI) score. In patients with PM, CD14(+) macrophages tended to be increased with enhanced expression of CD14, CD16 and a M2-macrophage marker, CD163. In particular, macrophages in patients with high TLR contained many granules with high side scatter and CD14 expression in their flow profile compared to those without PM. Conclusion: PM are accompanied by a drastic change in phenotypes of lymphocyte and macrophage in the peritoneal cavity, which might be involved in the development and progression of intraperitoneal tumor growth.


Subject(s)
Peritoneal Neoplasms , Stomach Neoplasms , CD8-Positive T-Lymphocytes/pathology , Humans , Killer Cells, Natural , Peritoneal Cavity , Peritoneal Neoplasms/secondary , Tumor Microenvironment
7.
Int J Biol Sci ; 18(11): 4400-4413, 2022.
Article in English | MEDLINE | ID: mdl-35864971

ABSTRACT

Endometriosis remains a common but challenging gynecological disease among reproductive-aged women with an unclear pathogenesis and limited therapeutic options. Numerous pieces of evidence suggest that NF-κB signaling, a major regulator of inflammatory responses, is overactive in endometriotic lesions and contributes to the onset, progression, and recurrence of endometriosis. Several factors, such as estrogen, progesterone, oxidative stress, and noncoding RNAs, can regulate NF-κB signaling in endometriosis. In the present review, we discuss the mechanisms by which these factors regulate NF-κB during endometriosis progression and provide an update on the role of NF-κB in affecting endometriotic cells, peritoneal macrophages (PMs) as well as endometriosis-related symptoms, such as pain and infertility. Furthermore, the preclinical drugs for blocking NF-κB signaling in endometriosis are summarized, including plant-derived medicines, NF-κB inhibitors, other known drugs, and the potential anti-NF-κB drugs predicted through the Drug-Gene Interaction Database. The present review discusses most of the studies concerning the multifaceted role of NF-κB signaling in endometriosis and provides a summary of NF-κB-targeted treatment in detail.


Subject(s)
Endometriosis , Adult , Estrogens , Female , Humans , NF-kappa B/metabolism , Signal Transduction/genetics
8.
Immunology ; 167(1): 77-93, 2022 09.
Article in English | MEDLINE | ID: mdl-35689656

ABSTRACT

The source of macrophages that contribute to human liver disease remains poorly understood. The purpose of this study is to investigate the functional mechanism of peritoneal macrophages in the development of hepatic immunopathology. By performing the natural infection with the blood fluke Schistosoma japonicum (S. japonicum) and the chemically carbon tetrachloride (CCl4 )-induced liver injured mouse model, we identified the peritoneal cavity as an essential source of hepatic macrophages. Here, we show that a large number of F4/80+ macrophages was accumulated in the peritoneal cavity during liver injury. An unknown source population of macrophages, which highly expressed GATA6 that is specific to peritoneal macrophages, was found to exist in the injured livers. Peritoneal macrophage deletion by injection with clodronate-containing liposomes led to an attenuated hepatic pathology and the inflammatory microenvironment, while adoptive transfer of macrophages into the abdominal cavity, by contrast, results in restoring liver pathology. Importantly, there are set genes of monocyte chemoattractant protein (MCP)-1, -2, and -3 that are highly related to recruit GATA6+ macrophages during S. japonicum infection, while administration of bindarit, a selective inhibitor of MCPs synthesis, dramatically decreased the hepatic expression of GATA6+ macrophages and thus attenuated hepatic pathology. Furthermore, in vivo study showed that peritoneal macrophages promote hepatic immunopathology is dependent on the accumulation of regulatory T cells (Tregs) in the liver. Altogether, these data provide the first clear evidence that GATA6+ peritoneal macrophages play critical roles in both the formation of hepatic immunopathology and the accumulation of Tregs cells.


Subject(s)
Schistosoma japonicum , Schistosomiasis japonica , Animals , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Humans , Liver/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Schistosomiasis japonica/metabolism , T-Lymphocytes, Regulatory/metabolism
9.
JHEP Rep ; 4(1): 100391, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34917912

ABSTRACT

BACKGROUND & AIMS: V-set Ig-domain-containing 4 (VSIG4) is an immunomodulatory macrophage complement receptor modulating innate and adaptive immunity and affecting the resolution of bacterial infections. Given its expression on peritoneal macrophages (PMs), we hypothesised a prognostic role of peritoneal VSIG4 concentrations in patients with spontaneous bacterial peritonitis (SBP). METHODS: We isolated PMs from patients with cirrhosis and analysed VSIG4 expression and release by flow cytometry, quantitative real-time PCR, ELISA, and confocal microscopy. We measured soluble VSIG4 concentrations in ascites from 120 patients with SBP and 40 patients without SBP and investigated the association of soluble VSIG4 in ascites with 90-day survival after SBP using Kaplan-Meier statistics, Cox regression, and competing-risks regression analysis. RESULTS: VSIG4 expression was high on resting, large PMs, which co-expressed CD206, CD163, and tyrosine-protein kinase Mer (MERTK). VSIG4 gene expression in PMs decreased in patients with SBP and normalised after resolution. During SBP, VSIG4hi PMs were depleted (25% vs. 57%; p <0.001) and soluble VSIG4 in ascites were higher in patients with SBP than in patients without (0.73 vs. 0.35 µg/ml; p <0.0001). PM activation by Toll-like receptor (TLR) agonists or infection with live bacteria in vitro resulted in a loss of surface VSIG4 and the release of soluble VSIG4. Mechanistically, shedding of VSIG4 from PMs was protease-dependent and susceptible to microtubule transport inhibition. Soluble VSIG4 in ascites exceeded serum concentrations and correlated with serum creatinine, model for end-stage liver disease score and C-reactive protein during SBP. Concentrations of 1.0206 µg/ml or higher indicated increased 90-day mortality (hazard ratio 1.70; 95% CI 1.01-2.86; p = 0.046). CONCLUSIONS: VSIG4 is released from activated PMs into ascites during SBP. Higher peritoneal VSIG4 levels indicate patients with organ failure and poor prognosis. LAY SUMMARY: Patients with liver cirrhosis who develop ascites have an increased risk of infection and mortality. Our study shows that in patients with infected ascites, the complement receptor VSIG4 is released by resident macrophages into the abdominal fluid where it can be measured. Patients with elevated levels of this protein in ascites are at high risk of dying within 90 days.

10.
Aging (Albany NY) ; 13(11): 15511-15522, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34111028

ABSTRACT

Propofol (Pro) confers protection against renal ischemia/reperfusion (rI/R) injury through incompletely characterized mechanisms. Since Pro has shown net anti-inflammatory properties as part of its beneficial effects, we examined the potential role of Pro in the modulation of macrophage polarization status during both rI/R injury in vivo and exposure of cultured peritoneal macrophages (PMs) to hypoxia/reoxygenation (H/R). Rats were subjected to 45-min r/IR surgery or a sham procedure and administered PBS (vehicle) or Pro during the ischemia stage. Pro administration attenuated rI/R-induced kidney damage and renal TNF-α, IL-6, and CXCL-10 expression. Enhanced macrophage M2 polarization, evidenced by reduced iNOS and increased Arg1 and Mrc1 mRNA levels, was further detected after Pro treatment both in the kidney, after rI/R in vivo, and in H/R-treated PMs. Pro administration also repressed phosphorylated signal transducer and activator of transcription 1 (p-STAT1) and increased p-STAT3, p-STAT6, and peroxisome proliferator-activated receptor-γ (PPARγ) mRNA levels in H/R-exposed PMs. Importantly, siRNA-mediated PPARγ silencing repressed Pro-mediated STAT3 activation in PMs and restored proinflammatory cytokine levels and prevented macrophage M2 marker expression in both rI/R-treated rats and cultured PMs. These findings suggest that Pro confers renoprotection against rI/R by stimulating PPARγ/STAT3-dependent macrophage conversion to the M2 phenotype.


Subject(s)
Cell Polarity , Macrophages/pathology , PPAR gamma/metabolism , Propofol/therapeutic use , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , STAT3 Transcription Factor/metabolism , Signal Transduction , Animals , Cell Polarity/drug effects , Gene Silencing/drug effects , Inflammation/pathology , Kidney/pathology , Macrophages/drug effects , Macrophages/metabolism , Male , Propofol/pharmacology , Protective Agents/pharmacology , Rats, Sprague-Dawley
11.
Parasitology ; 148(6): 703-711, 2021 05.
Article in English | MEDLINE | ID: mdl-33536085

ABSTRACT

Toxoplasma gondii can infect almost all warm-blooded vertebrates with pathogensis being largely influenced by the host immune status. As important epidemiological hosts, rodents are globally distributed and are also commonly found infected with haemoflagellates, such as those in the genus Trypanosoma. We here address whether and how co-infection with trypanosomes can influence T. gondii infection in laboratory models. Rats of five strains, co-infected with T. lewisi and mice of four strains, co-infected with T. musculi, were found to be more or less susceptible to T. gondii infection, respectively, with corresponding increased or decreased brain cyst burdens. Downregulation of iNOS expression and decreased NO production or reverse were observed in the peritoneal macrophages of rats or mice, infected with trypanosomes, respectively. Trypanosoma lewisi and T. musculi can modulate host immune responses, either by enhancement or suppression and influence the outcome of Toxoplasma infection.


Subject(s)
Toxoplasmosis/complications , Trypanosoma lewisi/physiology , Trypanosomiasis/complications , Animals , Blotting, Western , Brain/parasitology , Disease Models, Animal , Macrophages, Peritoneal , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Rats, Wistar , Specific Pathogen-Free Organisms , Splenomegaly , Toxoplasma/physiology , Toxoplasmosis/epidemiology , Trypanosoma/classification , Trypanosoma/physiology , Trypanosomiasis/immunology , Trypanosomiasis/parasitology
12.
Aging (Albany NY) ; 13(5): 7096-7119, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33639613

ABSTRACT

Atherosclerosis is a chronic inflammatory disease that commonly affects the elderly and is characterized by vascular damage, macrophage infiltration, and plaque formation. Moreover, it increases the risk of cardiovascular disease. The pathogenesis of atherosclerosis involves an interplay between macrophage autophagy and apoptosis. A recently discovered transcription factor, transcription factor EB (TFEB) is known to activate autophagy in macrophages. Sirtuin deacetylase 1 (SIRT1), a nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylase, activates several transcription factors, including TFEB. We studied the effects of berberine on the NAD+ synthesis pathway and interactions between SIRT1 and TFEB. We also studied the effects of berberine-induced TFEB activation via SIRT1 on autophagy and apoptosis of peritoneal macrophages. We found that berberine promoted autophagy of peritoneal macrophages by activating SIRT1 via the NAD+ synthesis pathway and, in turn, promoting TFEB nuclear translocation and deacetylation. The functional regulation of SIRT1 and TFEB by berberine could be exploited as a potential therapeutic strategy for atherosclerosis.


Subject(s)
Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Berberine/pharmacology , Macrophages, Peritoneal/drug effects , Sirtuin 1/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Flow Cytometry , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred C57BL , NAD/metabolism
13.
Chinese Pharmacological Bulletin ; (12): 550-555, 2021.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1014397

ABSTRACT

Aim To explore the anti-inflammatory effect of natural compound Ginkgetin on lipopolysaccharide-induced mouse primary peritoneal macrophages and the underlying mechanism, in order to provide a theoretical basis for the development of clinical drug candidates. Methods MTT test kit was used to detect the cytotoxicity of Ginkgetin on mouse primary peritoneal macrophages; ELISA and RT-qPCR methods were used to detect the anti-inflammatory effect of different concentrations of Ginkgetin on LPS-induced cell inflammation injury model; Western blot was used to detect the anti-inflammatory mechanism of ginkgo flavonoids. Results Compared with LPS stimulation group, Ginkgetin treatment group produced a concentration-dependent anti-inflammatory effect, which could be attributed to the fact that Ginkgetin could inhibit LPS-induced activation of NF-κB signaling pathway. MTT results also showed that ginkgo flavonoids had little toxicity to mouse primary peritoneal macrophages. Conclusions Ginkgelin alleviates LPS-induced inflammatory injury of mouse primary peritoneal macrophages by inhibiting the activation of NF-κB signaling pathway. It is expected to be a natural monomer antiinflammatory drug candidate.

14.
In Vitro Cell Dev Biol Anim ; 56(2): 154-164, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31898012

ABSTRACT

Cigarette smoke exposure is one of the main etiologies for chronic obstructive pulmonary disease. Moreover, cigarette smoke participates in disease progression by inducing abnormal macrophage polarization; however, the effects of cigarette smoke on M1/M2 macrophage polarization have not been established. The aim of the current study was to determine the effects of cigarette smoke extract (CSE) on M1/M2 macrophage polarization in alveolar and peritoneal macrophages (AM and PM, respectively) at different concentrations and exposure times. Rat AM and PM were cultured with CSE at different concentrations. CCK-8 was used as an indicator of cell viability, and mRNA expression of M1 (iNOS, TNF-α, and IL-1ß) and M2 markers (arg-1, CD206, and TGF-ß1) were measured at 3, 6, 9, 12, and 24 h using qPCR. Expressions of CD86 and CD206 proteins at 12 h were determined using flow cytometry, and the iNOS/arg-1 ratio was used to determine the polarization dominance of M1 and M2. M2 subtypes were detected at 12 h using qPCR and flow cytometry. CSE increased the expression of iNOS, TNF-α, and IL-1ß mRNA, and the proportion of CD86-positive cells in AM and PM promoted M1 polarization, and M1 polarization was continuously enhanced as exposure time and concentration increased. CSE reduced the expression of arg-1, CD206, and TGF-ß1 mRNA and the proportion of CD206-positive cells in AM and PM and inhibited M2 polarization. At 9-24 h of CSE exposure, the expression of arg-1 in AM and PM gradually increased, showing tendency towards activation of M2 polarization. Besides, CSE might induce M2b and M2d polarization at 12 h. After 12 h of CSE exposure, transformation from M1 to M2 polarization dominance was shown in AM; however, M1 polarization was continuously enhanced in PM within 24 h of CSE exposure. CSE promoted M1 polarization in macrophages, exhibiting dynamic regulatory effects on M2 polarization, first as a suppressor and then as a promoter. The polarization change induced by CSE on AM was more sensitive than PM.


Subject(s)
Cell Polarity , Cigarette Smoking/adverse effects , Macrophages, Alveolar/pathology , Macrophages, Peritoneal/pathology , Animals , Antigens, CD/metabolism , Arginase/metabolism , Cell Polarity/genetics , Cell Shape/genetics , Cell Survival/genetics , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Macrophages, Alveolar/metabolism , Macrophages, Peritoneal/metabolism , Male , Nitric Oxide Synthase Type II/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Time Factors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
15.
Pharm Biol ; 57(1): 485-497, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31401911

ABSTRACT

Context: Genus Verbascum (Scrophulariaceae) comprises about 360 species of flowering plants. Verbascum has been used in traditional medicine as an astringent, antitussive, analgesic and anti-inflammatory. Objective: Nothing was found in the available literature concerning Verbascum nubicum Murb; therefore, the study evaluates the biological activities, isolated compounds and HPLC profile. Materials and methods: Methanol extract (VME) and butanol fraction (VBF) of air-dried powdered V. nubicum were obtained. Four compounds were isolated from VBE and identified by 1H- and 13C-NMR. High-performance liquid chromatography (HPLC) profile was determined for (VME). LD50, in vitro antioxidant, in vivo antiulcerogenic and anti-inflammatory activities as well as hepatoprotective activity were assessed. Anti-ulcerogenic and hepatoprotective activities were supported by histopathological examinations. Results: HPLC analysis of VME revealed the presence of luteolin 7-glucoside (2215.43 mg/100 g), hesperidin (954.51 mg/100 g) and apigenin (233.15 mg/100 g) as major compounds. Four compounds were isolated and confirmed by NMR data, were identified as gentiopicroside, luteolin, aucubin and gallic acid. The LD50 of VME and VBF extracts were calculated to be 8200 and 4225 mg/kg b.w., respectively. IC50 values of VBE and VMF as measured by DPPH·method were 43.6 and 50 µg/mL, respectively. Also, anti-inflammatory effect of VME (400 mg/kg b.w.) and VBF (200 mg/kg b.w.) induced edema model after 120 min were 61.93 and 56.13%, respectively. Antiulcerogenic activity of VME (400 mg/kg b.w.) and VBF (200 mg/kg b.w.) in albino rats were 65.14 and 84.57%, respectively. Conclusions: The V. nubicum extracts displayed safe and promising antioxidant, anti-inflammatory and hepatoprotective properties. It can be also applied in the pharmacy industry, food industry and healthcare.


Subject(s)
Plant Extracts/pharmacology , Verbascum/chemistry , Analgesics/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Apigenin/analysis , Chromatography, High Pressure Liquid , Edema/chemically induced , Edema/drug therapy , Glucosides/analysis , Hesperidin/analysis , Lethal Dose 50 , Luteolin/analysis , Medicine, Traditional , Plant Extracts/toxicity , Rats , Stomach Ulcer/chemically induced , Stomach Ulcer/drug therapy , Stomach Ulcer/pathology
16.
Immunol Res ; 67(2-3): 241-260, 2019 06.
Article in English | MEDLINE | ID: mdl-31290001

ABSTRACT

Our earlier studies proposed a radically new idea suggesting interdependency between TNF-α/TNFR1 and IL-1ß/IL-1R pathways in modulation of Staphylococcus aureus-induced CXCL8/CXCR1 axis. However, the effects of inhibition of cytokine receptor mobilization at intracellular level and surface TNFR1 and IL-1R shedding on S. aureus-induced CXCR1 expression have not been studied so far in peritoneal macrophages. This study aimed to investigate the role of inhibition of receptor mobilization from the intracellular pool (using brefeldin A) and surface receptor shedding (using TAPI-1) on CXCR1 expression during dual receptor (TNFR1 plus IL-1R) neutralization in peritoneal macrophages isolated from wild-type Swiss Albino mice. Release of superoxide anion, nitric oxide, and hydrogen peroxide was measured and cytokine production was done by ELISA. Expression of surface receptors (TNFR1, IL-1R, and CXCR1) and inflammatory mediators was studied by Western blot. It was observed that S. aureus-infected macrophages showed elevated ROS production, secretion of TNF-α, IL-1ß, and CXCL8, along with increased expression of surface receptors (TNFR1, IL-1R, and CXCR1), and inflammatory markers (iNOS and COX-2) compared with control or treated groups (p < 0.05). However, prior treatment of macrophages with BFA or TAPI-1 in the presence of anti-TNFR1 antibody and IRAP during S. aureus infection showed significant reduction of all these parameters (p < 0.05). We can conclude that targeting of TNFR1 and IL-1R (with major focus on surface expression study) either through blockage of intracellular receptor trafficking pathway or via surface receptor shedding diminishes TNFR1/IL-1R interaction and consequently downregulates CXCR1 expression along with inflammatory signalling pathways during bacterial infections.


Subject(s)
Host-Pathogen Interactions , Receptors, Interleukin-1 Type I/metabolism , Receptors, Interleukin-8A/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcus aureus/immunology , Biomarkers , Cytokines/metabolism , Humans , Hydrogen Peroxide/metabolism , Nitric Oxide/metabolism , Receptors, Interleukin-8A/genetics , Staphylococcal Infections/genetics
17.
Eur J Immunol ; 49(11): 2012-2018, 2019 11.
Article in English | MEDLINE | ID: mdl-31251389

ABSTRACT

Macrophages play a central role during infection, inflammation and tissue homeostasis maintenance. Macrophages have been identified in all organs and their core transcriptomic signature and functions differ from one tissue to another. Interestingly, macrophages have also been identified in the peritoneal cavity and these cells have been extensively used as a model for phagocytosis, efferocytosis and polarization. Peritoneal macrophages are involved in B-cell IgA production, control of inflammation and wound healing following thermal-induced liver surface injury. These cells presumably require and interact with the omentum, where milky spot stromal cells have been proposed to secrete CSF1 (colony stimulating factor 1). Peritoneal macrophages depend on CSF1 for their generation and survival, but the identity of CSF1 producing cells inside the large peritoneal cavity remains unknown. Here we investigated peritoneal macrophage localization and their interaction with mesothelial cells, the major cell type predicted to secrete CSF1. Our data revealed that mesothelial cells produce membrane bound and secreted CSF1 that both sustain peritoneal macrophage growth.


Subject(s)
Epithelial Cells/metabolism , Epithelium/metabolism , Macrophage Colony-Stimulating Factor/genetics , Macrophages, Peritoneal/metabolism , Stromal Cells/metabolism , Animals , Cell Communication/genetics , Cell Communication/immunology , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Survival , Coculture Techniques , Epithelial Cells/cytology , Epithelial Cells/immunology , Epithelium/immunology , Extracellular Space/immunology , Extracellular Space/metabolism , Gene Expression , Macrophage Colony-Stimulating Factor/immunology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/immunology , Mice , Mice, Transgenic , Peritoneal Cavity/cytology , Signal Transduction , Stromal Cells/cytology , Stromal Cells/immunology
18.
Cell Physiol Biochem ; 52(6): 1325-1338, 2019.
Article in English | MEDLINE | ID: mdl-31050281

ABSTRACT

BACKGROUND/AIMS: Atherosclerosis is a chronic inflammatory cardiovascular disease. Macrophages are major components of atherosclerotic plaques and play a key role in the development of atherosclerosis by secreting a variety of pro-inflammatory factors. Our previous studies have confirmed that upconversion nanoparticles encapsulating chlorin e6 (UCNPs-Ce6) mediated photodynamic therapy (PDT) can promote cholesterol efflux and induce apoptosis in THP-1 macrophages. In this study, we investigated whether reactive oxygen species (ROS) generated by UCNPs-Ce6-mediated PDT can induce autophagy to inhibit the expression of pro-inflammatory factor in M1 peritoneal macrophages. METHODS: Peritoneal macrophages were collected from C57/BL6 mice injected with 3% thioglycollate broth medium and induced by lipopolysaccharides and interferon-γ. Intracellular ROS production was assessed by 2'-7'-dichloroflorescein diacetate and flow cytometry. Autophagy was assayed by western blot, transmission electron microscopy and immunofluorescence. Pro-inflammatory cytokines were detected by enzyme-linked immunosorbent assay and western blot. RESULTS: Model M1 peritoneal macrophages were established after 24 h induction. Protein expression levels of LC3 II and Beclin1, and degradation of p62 increased and peaked at 2 h in the PDT group. Meanwhile, levels of inflammatory cytokines iNOS, IL-12, and TNF-α markedly decreased after PDT. The increase in autophagy levels and decrease in pro-inflammatory cytokines were significantly inhibited by 3-methyladenine. Furthermore, ROS generated by UCNPs- Ce6 mediated PDT activated autophagy. The expression of autophagy related-protein and inflammatory cytokines iNOS, IL-12, and TNF-α were inhibited by the ROS inhibitor N-acetyl cysteine. CONCLUSION: ROS generated by UCNPs-Ce6-mediated PDT activated autophagy and inhibited the expression of pro-inflammatory factors of M1 peritoneal macrophage via the PI3K/AKT/mTOR signaling pathway.


Subject(s)
Autophagy/drug effects , Metal Nanoparticles/chemistry , Porphyrins/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Beclin-1/metabolism , Interleukin-12/metabolism , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Nitric Oxide Synthase Type II/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Photochemotherapy , Porphyrins/chemistry , Porphyrins/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism
19.
Int Immunopharmacol ; 69: 178-183, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30716588

ABSTRACT

Glucose-6-phosphate dehydrogenase (G6PDH) ultimately plays a critical role in macrophage functions used against infectious agents. The present study investigated whether changes in G6PDH activity could influence the resistance of infected macrophages against Leishmania major infection. Mouse peritoneal and J774 macrophages were infected, respectively, ex vivo and in vitro, with L. major and then exposed to an inhibitor (6-aminonicotinamide) or activator (LPS + melatonin) of G6PDH activity for 24 h. Cell viability [using MTT assay] was measured to assess any direct toxicity from the doses of inhibitor/activator used for the macrophage treatments. Nitric oxide (NO) produced by the cells and released into culture supernatants was measured (Griess method) and cell G6PDH activity was also determined. Moreover, the number of amastigotes form Leishmania in macrophages that developed over a 7-d period was evaluated. The results showed that an increase in G6PDH activity after treatment of both types of macrophages with a combination of LPS + melatonin caused significant increases in NO production and cell resistance against L. major amastigote formation/survival. However, exposure to 6-aminonicotinamide led to remarkable suppression of G6PDH activity and NO production, events that were associated with a deterioration in cell resistance against (and an increase in cell levels of) the parasites. The results suggested that activation or suppression of G6PDH activity could affect leishmanicidal function of both mouse peritoneal and J774 macrophages. Thus, regulation of macrophages via modulation of G6PDH activity appears to provide a novel window for those seeking to develop alternative therapies for the treatment of leishmaniasis.


Subject(s)
Glucosephosphate Dehydrogenase/metabolism , Leishmania major/physiology , Macrophages, Peritoneal/physiology , Animals , Cell Line , Disease Models, Animal , Humans , Immunomodulation , Leishmaniasis, Cutaneous , Macrophages, Peritoneal/parasitology , Mice , Mice, Inbred BALB C , Nitric Oxide/metabolism , Oxidation-Reduction
20.
Cell Physiol Biochem ; 48(4): 1616-1627, 2018.
Article in English | MEDLINE | ID: mdl-30071509

ABSTRACT

BACKGROUND/AIMS: Atherosclerosis is a chronic inflammatory cardiovascular disease. Macrophages are major components of atherosclerotic plaques and play a key role in the development of atherosclerosis by secreting a variety of pro-inflammatory factors. Our previous studies have confirmed that upconversion nanoparticles encapsulating chlorin e6 (UCNPs-Ce6) mediated photodynamic therapy (PDT) can promote cholesterol efflux and induce apoptosis in THP-1 macrophages. In this study, we investigated whether reactive oxygen species (ROS) generated by UCNPs-Ce6-mediated PDT can induce autophagy to inhibit the expression of pro-inflammatory factor in M1 peritoneal macrophages. METHODS: Peritoneal macrophages were collected from C57/BL6 mice injected with 3% thioglycollate broth medium and induced by lipopolysaccharides and interferon-γ. Intracellular ROS production was assessed by 2'-7'-dichloroforescein diacetate and flow cytometry. Autophagy was assayed by western blot, transmission electron microscopy and immunofluorescence. Pro-inflammatory cytokines were detected by enzyme-linked immunosorbent assay and western blot. RESULTS: Model M1 peritoneal macrophages were established after 24 h induction. Protein expression levels of LC3 II and Beclin1, and degradation of p62 increased and peaked at 2 h in the PDT group. Meanwhile, levels of inflammatory cytokines iNOS, IL-12, and TNF-α markedly decreased after PDT. The increase in autophagy levels and decrease in pro-inflammatory cytokines were significantly inhibited by 3-methyladenine. Furthermore, ROS generated by UCNPs-Ce6 mediated PDT activated autophagy. The expression of autophagy related-protein and inflammatory cytokines iNOS, IL-12, and TNF-α were inhibited by the ROS inhibitor N-acetyl cysteine. CONCLUSIONS: ROS generated by UCNPs-Ce6-mediated PDT activated autophagy and inhibited the expression of pro-inflammatory factors of M1 peritoneal macrophage via the PI3K/AKT/mTOR signaling pathway.


Subject(s)
Autophagy/drug effects , Magnetite Nanoparticles/toxicity , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Animals , Cell Survival/drug effects , Enzyme Inhibitors/pharmacology , Interleukin-12/metabolism , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Magnetite Nanoparticles/chemistry , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Nitric Oxide Synthase Type II/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Photochemotherapy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Receptors, IgG/metabolism , TOR Serine-Threonine Kinases/metabolism , Transcription Factor TFIIH , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL