Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Development ; 151(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38884383

ABSTRACT

The specialized cell cycle of meiosis transforms diploid germ cells into haploid gametes. In mammals, diploid spermatogenic cells acquire the competence to initiate meiosis in response to retinoic acid. Previous mouse studies revealed that MEIOC interacts with RNA-binding proteins YTHDC2 and RBM46 to repress mitotic genes and to promote robust meiotic gene expression in spermatogenic cells that have initiated meiosis. Here, we have used the enhanced resolution of scRNA-seq and bulk RNA-seq of developmentally synchronized spermatogenesis to define how MEIOC molecularly supports early meiosis in spermatogenic cells. We demonstrate that MEIOC mediates transcriptomic changes before meiotic initiation, earlier than previously appreciated. MEIOC, acting with YTHDC2 and RBM46, destabilizes its mRNA targets, including the transcriptional repressors E2f6 and Mga, in mitotic spermatogonia. MEIOC thereby derepresses E2F6- and MGA-repressed genes, including Meiosin and other meiosis-associated genes. This confers on spermatogenic cells the molecular competence to, in response to retinoic acid, fully activate the transcriptional regulator STRA8-MEIOSIN, which is required for the meiotic G1/S phase transition and for meiotic gene expression. We conclude that, in mice, mRNA decay mediated by MEIOC-YTHDC2-RBM46 enhances the competence of spermatogenic cells to initiate meiosis.


Subject(s)
Meiosis , RNA, Messenger , RNA-Binding Proteins , Spermatogenesis , Animals , Male , Mice , RNA, Messenger/metabolism , RNA, Messenger/genetics , Spermatogenesis/genetics , Spermatogenesis/physiology , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Spermatogonia/metabolism , Spermatogonia/cytology , Tretinoin/metabolism , Tretinoin/pharmacology , RNA Stability/genetics , Gene Expression Regulation, Developmental , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , RNA Helicases
2.
bioRxiv ; 2023 Sep 22.
Article in English | MEDLINE | ID: mdl-37781613

ABSTRACT

The special cell cycle known as meiosis transforms diploid germ cells into haploid gametes. In mammalian testes, diploid spermatogenic cells become competent to transition from mitosis to meiosis in response to retinoic acid. In mice, previous studies revealed that MEIOC, alongside binding partners YTHDC2 and RBM46, represses mitotic genes and promotes robust meiotic gene expression in spermatogenic cells that have already initiated meiosis. Here, we molecularly dissect MEIOC-dependent regulation in mouse spermatogenic cells and find that MEIOC actually shapes the transcriptome much earlier, even before meiotic initiation. MEIOC, acting with YTHDC2 and RBM46, destabilizes mRNA targets, including transcriptional repressors E2f6 and Mga, in mitotic spermatogonia. MEIOC thereby derepresses E2F6- and MGA-repressed genes, including Meiosin and other meiosis-associated genes. This confers on spermatogenic cells the molecular competence to, in response to retinoic acid, fully activate the STRA8-MEIOSIN transcriptional regulator, which is required for the meiotic G1/S cell cycle transition and meiotic gene expression. We conclude that in mice, mRNA decay mediated by MEIOC-YTHDC2-RBM46 enhances the competence of mitotic spermatogonia to transit from mitosis to meiosis.

4.
Front Immunol ; 10: 2650, 2019.
Article in English | MEDLINE | ID: mdl-31798583

ABSTRACT

Microglia are the brain's immune cells and play an important role in regulating the microenvironment in the central nervous system. Activated microglia are capable of acquiring the pro-inflammatory (M1) phenotype and anti-inflammatory (M2) phenotype. Overactivation of microglia is neurotoxic and may lead to neuroinflammatory brain disorders. Neuroinflammation in the brain plays a crucial role part in the pathophysiology of many psychiatric and neurological diseases. The inhibition of M1 microglia and promotion of M2 microglia was demonstrated to treat and prevent these diseases through reduced neuroinflammation. Isovitexin (IVX) has anti-inflammatory properties and passes through the blood-brain barrier; however, the molecular mechanism that modulates IVX-mediated microglial polarization remains unclear. In BV-2 cells and mouse primary microglia, IVX suppressed the expression of M1 microglial markers, enhanced the expression of M2 microglial markers, and enhanced the release of interleukin 10 (IL-10). IVX promoted the expression of peroxisome proliferator-activated receptor-γ (PPARγ) and PPARγ coactivator-1α (PGC-1α) in LPS-induced microglial activation. The inhibition of PPARγ and PGC-1α attenuated the regulatory effect of IVX in LPS-induced microglial polarization. IVX increased the expression of p-CaMKKß, p-AMPK, and PGC-1α in BV-2 cells. Inhibition of CaMKKß with STO-609 or knockdown of CaMKKß with CaMKKß siRNA attenuated IVX-mediated M2 microglial polarization in LPS-treated cells. In LPS-treated mice, the inhibition of CaMKKß and PGC-1α attenuated the IVX-mediated prevention of sickness behavior and enhanction of IVX-mediated M2 microglial polarization. IVX promoted M2 microglial polarization which exerted anti-inflammatory effects on LPS-induced neuroinflammation via the activation of the CaMKKß/AMPK-PGC-1α signaling axis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Apigenin/pharmacology , Cell Differentiation/drug effects , Microglia/drug effects , Signal Transduction/drug effects , Adenylate Kinase/immunology , Adenylate Kinase/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Kinase/immunology , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Cell Differentiation/immunology , Inflammation/chemically induced , Inflammation/immunology , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Microglia/immunology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/immunology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Signal Transduction/immunology
5.
Front Immunol ; 10: 287, 2019.
Article in English | MEDLINE | ID: mdl-30858849

ABSTRACT

Background/Aims: Mastitis is an acute clinical inflammatory response. The occurrence and development of mastitis seriously disturb women's physical and mental health. Licochalcone A, a phenolic compound in Glycyrrhiza uralensis, has anti-inflammatory properties. Here, we examined the effect of licochalcone A on blood-milk barrier and inflammatory response in LPS-induced mice mastitis. Methods:In vivo, we firstly established mice models of mastitis by canal injection of LPS to mammary gland, and then detected the effect of licochalcone A on pathological indexes, inflammatory responses and blood-milk barrier in this model. In vivo, Mouse mammary epithelial cells (mMECs) were treated with licochalcone A prior to the incubation of LPS, and then the inflammatory responses, tight junction which is the basic structure of blood-milk barrier were analyzed. Last, we elucidated the anti-inflammatory mechanism by examining the activation of mitogen-activated protein kinase (MAPK) and AKT/NF-κB signaling pathways in vivo and in vitro. Result: The in vivo results showed that licochalcone A significantly decreased the histopathological impairment and the inflammatory responses, and improved integrity of blood-milk barrier. The in vitro results demonstrated that licochalcone A inhibited LPS-induced inflammatory responses and increase the protein levels of ZO-1, occludin, and claudin3 in mMECs. The in vivo and in vitro mechanistic study found that the anti-inflammatory effect of licochalcone A in LPS-induced mice mastitis was mediated by MAPK and AKT/NF-κB signaling pathways. Conclusions and Implications: Our experiments collectively indicate that licochalcone A protected against LPS-induced mice mastitis via improving the blood-milk barrier integrity and inhibits the inflammatory response by MAPK and AKT/NF-κB signaling pathways.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Chalcones/therapeutic use , Mastitis/drug therapy , Milk/metabolism , Animals , Cells, Cultured , Chalcones/pharmacology , Female , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/drug effects , Mastitis/pathology , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/physiology , Peroxidase/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/drug effects , Tight Junction Proteins/analysis
6.
J Cell Physiol ; 234(9): 16252-16262, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30746687

ABSTRACT

Mastitis, an inflammation of mammary gland, is a serious disease that affects the health of dairy cows around the world. Myricetin, a flavonoid from Bayberry, has been reported to suppress various inflammatory response. The aim of this study was to evaluate the effect of myricetin on lipopolysaccharide (LPS)-induced in vivo and in vitro mastitis model and clarify the underlying mechanism. In vivo experiments, myricetin attenuated the severity of inflammatory lesion and neutrophil infiltration. Moreover, myricetin pretreatment induced a significant decrease in the activity of myeloperoxidase (MPO) and the production of TNF-α, IL-6, and IL-1ß triggered by LPS. Myricetin pretreatment could also increase the integrity of the blood-milk barrier and upregulate the tight junction proteins in LPS-induced mice mastitis. In vitro, myricetin inhibited LPS-induced inflammatory response in mice mammary epithelial cells (mMECs). In the further mechanism studies, we found that the anti-inflammatory effect of myricetin was mediated by inhibiting LPS-induced phosphorylation of AKT, IKK-α, IκB-α, and P65 in vivo and in vitro. Collectively, these data suggested that myricetin effectively ameliorated the inflammatory response by inhibiting the AKT/IKK/NF-κB signaling pathway and repairing the integrity of blood-milk barrier in LPS-induced mice mastitis.

7.
Toxicol Appl Pharmacol ; 365: 9-18, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30610879

ABSTRACT

Vanillin is used in a variety of food, chemical, and pharmaceutical applications, and exhibits anti-inflammatory properties. However, there are no reports about the effects of vanillin on lipopolysaccharide (LPS)-induced mastitis. In this study, we explored the effects of vanillin on the subsequent inflammatory response and blood-milk barrier in LPS-induced mastitis. Results showed that vanillin suppressed the inflammatory response by a) inhibiting myeloperoxidase activity; b) decreasing the production of pro-inflammatory mediators which include tumor necrosis factor alpha (Tnf-α; from 128.5 ±â€¯14.59 to 67.51 ±â€¯10.88,pg/mL, P < 0.01), interleukin-6 (Il-6; from 531.5 ±â€¯196.4 to 109.3 ±â€¯24.14, pg/mL, P < 0.05), interleukin-1ß (Il-1ß; from 2569 ±â€¯1648 to 731.8 ±â€¯171.7, pg/mL, P < 0.05), inducible nitric oxide synthase (Inos), and cyclooxygenase-2 (Cox-2); and c) repairing the blood-milk barrier by increasing the protein levels of the tight junction proteins, including zona occludens 1 (Zo-1), claudin-3, and occludin. In vitro experiment, Vanillin can inhibit LPS-induced inflammation and enhance the protein levels of tight junction proteins. Further studies have shown that vanillin inhibits inflammation by inhibiting mitogen-activated protein kinases (MAPKs) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathways. Our findings showed that vanillin protects mammary gland from LPS-induced mastitis by enhancing the blood-milk barrier and inhibiting the inflammatory response.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Benzaldehydes/pharmacology , Epithelial Cells/drug effects , Inflammation Mediators/metabolism , Lipopolysaccharides , Mammary Glands, Animal/drug effects , Mastitis/drug therapy , Animals , Cells, Cultured , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/metabolism , Female , Inflammation Mediators/immunology , Lactation , Mammary Glands, Animal/immunology , Mammary Glands, Animal/metabolism , Mastitis/chemically induced , Mastitis/immunology , Mastitis/metabolism , Mice, Inbred BALB C , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Peroxidase/metabolism , Pregnancy , Signal Transduction/drug effects , Tight Junction Proteins/metabolism , Tight Junctions/drug effects , Tight Junctions/metabolism
8.
PLoS One ; 12(4): e0173754, 2017.
Article in English | MEDLINE | ID: mdl-28399128

ABSTRACT

Research on human immunology has been hindered by the lack of optimal small animal models, given that the protective immune responses of human and non-human species show significant differences. However, due to ethical constraints[1] and the high cost of clinical trials, it is urgent to improve the current animal models that can mimic faithfully human physiology, particularly the human immune system (HIS). HIS mice had been generated recently by engrafting human hematopoietic stem cells (hHSCs) or human peripheral mononuclear cells (hPBMCs) into highly immuno-deficient mice such as NSG, NOG or NRG mice. However, a major experimental drawback for studies using these models is the rapid onset of Graft-versus-Host Disease (GvHD). In the present study, we overcome this limitation by generating new immuno-deficient mice named "HUMAMICE" (HLA-A2+/+/DR1+/+/H-2-ß2m-/-/IAß-/-/Rag2-/-/IL2rγ-/-/Perf-/- mice), which expressed human HLA molecules instead of mouse MHC molecules (H-2), and whose immuno-deficient status was reversed by transferring functional HLA-matched PBMCs thus producing mice with an immuno-competent status with a functional human immune system. We showed that in this HLA-matched context, the hPBMC-transfer led to high lymphocytes engraftment rates without GvHD over three months in this novel mouse model. Furthermore, to evaluate the utility of the hPBMC-HUMAMICE, we immunized them with commercial vaccine of Hepatitis B virus (HBsAg, Hepvac@) which resulted in robust and reproducible production of high levels of HBsAg-specific antibodies, implying that both transferred T and B lymphocytes were functional in HUMAMICE. These responses are comparable to those observed in human clinical trials with this identical vaccine. In conclusion, these findings indicated that the HLA-matched-hPBMC-HUMAMICE represents a promising model for dissecting human immune responses in various human diseases, including infectious diseases, cancers and tumors, and to facilitate the development of novel vaccines and cellular therapies.


Subject(s)
HLA-A2 Antigen , Hepatitis B Antibodies/biosynthesis , Mice, Transgenic , Models, Animal , Animals , Cell Line, Tumor , Female , Graft vs Host Disease , HLA-A2 Antigen/genetics , Hepatitis B Surface Antigens/immunology , Hepatitis B Vaccines/immunology , Humans , Immunologic Deficiency Syndromes , Leukocytes, Mononuclear/transplantation , Lymphocytes/immunology , Major Histocompatibility Complex , Mice, Inbred C57BL , Neoplasm Transplantation , Spleen/cytology , Spleen/metabolism , Vaccination
9.
In Vitro Cell Dev Biol Anim ; 52(3): 296-304, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26744030

ABSTRACT

The aim of this study was to establish a bovine anterior pituitary-derived lactotroph (BAPDL) line that expresses prolactin (PRL) in vitro to study the mechanisms of bovine PRL synthesis and secretion. Immunohistochemistry assay of PRL in the newborn calves' anterior pituitary glands showed that most lactotrophs were located within the superior border of the lateral wings of the anterior pituitary. Tissues of the superior border of the lateral wings of the anterior pituitary were dispersed and cultured in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS). The limiting dilution method was used to establish BAPDL from single cell clone. BAPDL cells constantly expressed mRNAs for PRL and pituitary-specific transcription factor 1 (Pit-1) gene and grew steadily and rapidly in the DMEM supplemented with 10% FBS. PRL immunoreactivity was present in BAPDL at passage 20. The concentration of bovine PRL in BAPDL at passage 20 culture supernatant was decreased to below 35% compared with that in BAPDL at passage 1. The effects of human epidermal growth factor (hEGF) and dopamine (DA) on the expression and secretion of PRL in BAPDL at passage 4 were also investigated. The results are consistent with those of previous studies. Thus, it can be used successfully for studying the mechanisms of stimuli regulating PRL synthesis and release.


Subject(s)
Dopamine/pharmacology , Epidermal Growth Factor/pharmacology , Lactotrophs/metabolism , Pituitary Gland, Anterior/metabolism , Prolactin/biosynthesis , Transcription Factor Pit-1/biosynthesis , Animals , Cattle , Cells, Cultured , Lactotrophs/cytology , Pituitary Gland, Anterior/cytology , Prolactin/genetics , Prolactin/metabolism , RNA, Messenger/biosynthesis , Transcription Factor Pit-1/genetics
10.
Front Microbiol ; 6: 1031, 2015.
Article in English | MEDLINE | ID: mdl-26528244

ABSTRACT

The emergence and wide spread of multi-drug resistant Staphylococcus aureus (S. aureus) requires the development of new therapeutic agents with alternative modes of action. Anti-virulence strategies are hoped to meet that need. Sortase A (SrtA) has attracted great interest as a potential drug target to treat infections caused by S. aureus, as many of the surface proteins displayed by SrtA function as virulence factors by mediating bacterial adhesion to specific organ tissues, invasion of host cells, and evasion of the host-immune responses. It has been suggested that inhibitors of SrtA might be promising candidates for the treatment and/or prevention of S. aureus infections. In this study, we report that chlorogenic acid (CHA), a natural compound that lacks significant anti-S. aureus activity, inhibit the activity of SrtA in vitro (IC50 = 33.86 ± 5.55 µg/ml) and the binding of S. aureus to fibrinogen (Fg). Using molecular dynamics simulations and mutagenesis assays, we further demonstrate that CHA binds to the binding sites of C184 and G192 in the SrtA. In vivo studies demonstrated that CHA prevent mice from S. aureus-induced renal abscess, resulting in a significant survival advantage. These findings indicate that CHA is a promising therapeutic compound against SrtA during S. aureus infections.

11.
Pathog Dis ; 73(6): ftv042, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26054573

ABSTRACT

Sortase A (SrtA) is a cysteine transpeptidase and virulence factor from Staphylococcus aureus (S. aureus) that catalyses the attachment and display of surface proteins on the cell wall, thereby mediating bacterial adhesion to host tissues, host-cell entry and evasion of the immune response. As a result, SrtA has become an important target in the development of therapies for S. aureus infections. In this study, we used the new reference strain S. aureus Newman D2C to investigate the role of SrtA in a murine model of bloodstream infection, when the impact of coagulase and haemolysin is excluded. The results suggested that deletion of SrtA reduced the bacterial burden on the heart, liver and kidneys by blunting the host proinflammatory cytokine response at an early point in infection. Kidneys, but not heart or liver, formed abscesses on the sixth day following non-lethal infection, and this effect was diminished by SrtA mutation. These findings indicate that SrtA is a determining virulence factor in lethality and formation of renal abscesses in mice followed by S. aureus bloodstream infection. We have thus established a convenient in vitro and mouse model for developing SrtA-targeted therapeutic strategies.


Subject(s)
Aminoacyltransferases/metabolism , Bacteremia/microbiology , Bacterial Proteins/metabolism , Coagulase/deficiency , Cysteine Endopeptidases/metabolism , Hemolysin Proteins/deficiency , Staphylococcus aureus/growth & development , Virulence Factors/metabolism , Abscess/microbiology , Abscess/pathology , Aminoacyltransferases/deficiency , Animals , Bacteremia/pathology , Bacterial Load , Cysteine Endopeptidases/deficiency , Disease Models, Animal , Female , Gene Deletion , Heart/microbiology , Kidney/microbiology , Kidney/pathology , Liver/microbiology , Liver/pathology , Mice, Inbred BALB C , Myocardium/pathology , Staphylococcus aureus/genetics , Survival Analysis , Virulence Factors/deficiency
12.
Molecules ; 20(4): 6533-43, 2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25871372

ABSTRACT

Sortase A (SrtA) is a cysteine transpeptidase of most Gram-positive bacteria that is responsible for the anchorage of many surface protein virulence factors to the cell wall layer. SrtA mutants are unable to display surface proteins and are defective in the establishment of infections without affecting microbial viability. In this study, we report that quercitrin (QEN), a natural compound that does not affect Staphylococcus aureus growth, can inhibit the catalytic activity of SrtA in fibrinogen (Fg) cell-clumping and immobilized fibronectin (Fn) adhesion assays. Molecular dynamics simulations and mutagenesis assays suggest that QEN binds to the binding sites of the SrtA G167A and V193A mutants. These findings indicate that QEN is a potential lead compound for the development of new anti-virulence agents against S. aureus infections.


Subject(s)
Aminoacyltransferases/antagonists & inhibitors , Bacterial Adhesion/drug effects , Bacterial Proteins/antagonists & inhibitors , Quercetin/analogs & derivatives , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Aminoacyltransferases/chemistry , Aminoacyltransferases/metabolism , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites , Catalysis/drug effects , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Microbial Sensitivity Tests , Models, Molecular , Molecular Conformation , Protein Binding , Quercetin/chemistry , Quercetin/pharmacology
13.
Int J Mol Sci ; 16(2): 4265-80, 2015 Feb 16.
Article in English | MEDLINE | ID: mdl-25690038

ABSTRACT

ß-hydroxybutyric acid (BHBA) regulates the synthesis and secretion of growth hormone (GH) and prolactin (PRL), but its mechanism is unknown. In this study, we detected the effects of BHBA on the activities of G protein signaling pathways, AMPK-α activity, GH, and PRL gene transcription, and GH and PRL secretion in dairy cow anterior pituitary cells (DCAPCs). The results showed that BHBA decreased intracellular cAMP levels and a subsequent reduction in protein kinase A (PKA) activity. Inhibition of PKA activity reduced cAMP response element-binding protein (CREB) phosphorylation, thereby inhibiting GH and PRL transcription and secretion. The effects of BHBA were attenuated by a specific Gαi inhibitor, pertussis toxin (PTX). In addition, intracellular BHBA uptake mediated by monocarboxylate transporter 1 (MCT1) could trigger AMPK signaling and result in the decrease in GH and PRL mRNA translation in DCAPCs cultured under low-glucose and non-glucose condition when compared with the high-glucose group. This study identifies a biochemical mechanism for the regulatory action of BHBA on GH and PRL gene transcription, translation, and secretion in DCAPCs, which may be one of the factors that regulate pituitary function during the transition period in dairy cows.


Subject(s)
Growth Hormone/metabolism , Hydroxybutyrates/toxicity , Pituitary Gland, Anterior/drug effects , Prolactin/metabolism , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Cattle , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Glucose/pharmacology , Growth Hormone/antagonists & inhibitors , Growth Hormone/genetics , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Pertussis Toxin/pharmacology , Phosphorylation/drug effects , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/metabolism , Prolactin/antagonists & inhibitors , RNA, Messenger/metabolism , Symporters/genetics , Symporters/metabolism , Transcription Factor Pit-1/genetics , Transcription Factor Pit-1/metabolism , Transcription, Genetic/drug effects
14.
J Neuroinflammation ; 12: 9, 2015 Jan 17.
Article in English | MEDLINE | ID: mdl-25595674

ABSTRACT

BACKGROUND: Accumulating evidence suggests that neuroinflammation plays an important role in the progression of Parkinson's disease (PD). Excessively activated microglia produce several pro-inflammatory enzymes and pro-inflammatory cytokines, leading to damage to surrounding neurons and eventually inducing neurodegeneration. Therefore, the inhibition of microglial overactivation may be a potential therapeutic strategy to prevent the further progression of PD. ß-Hydroxybutyric acid (BHBA) has been shown to suppress lipopolysaccharide (LPS)-induced inflammation in BV-2 cells and to protect dopaminergic neurons in previous studies, but the underlying mechanisms remain unclear. Thus, in this study, we further investigated this mechanism in LPS-induced in vivo and in vitro PD models. METHODS: For the in vitro experiments, primary mesencephalic neuron-glia cultures were pretreated with BHBA and stimulated with LPS. [(3)H]dopamine (DA) uptake, tyrosine hydroxylase-immunoreactive (TH-ir) neurons and morphological analysis were evaluated and analyzed in primary mesencephalic neuron-glia cultures. In vivo, microglial activation and the injury of dopaminergic neurons were induced by LPS intranigral injection, and the effects of BHBA treatment on microglial activation and the survival ratio and function of dopaminergic neurons were investigated. Four our in vitro mechanistic experiment, primary microglial cells were pretreated with BHBA and stimulated with LPS; the cells were then assessed for the responses of pro-inflammatory enzymes and pro-inflammatory cytokines, and the NF-κB signaling pathway was evaluated and analyzed. RESULTS: We found that BHBA concentration-dependently attenuated the LPS-induced decrease in [(3)H]DA uptake and loss of TH-ir neurons in the primary mesencephalic neuron/glia mixed culture. BHBA treatment significantly improved the motor dysfunction of the PD model rats induced by intranigral injection of LPS, and this beneficial effect of BHBA was attributed to the inhibition of microglial overactivation and the protection of dopaminergic neurons in the substantia nigra (SN). Our in vitro mechanistic study revealed that the inhibitory effect of BHBA on microglia was mediated by G-protein-coupled receptor 109A (GPR109A) and involved the NF-κB signaling pathway, causing the inhibition of pro-inflammatory enzyme (iNOS and COX-2) and pro-inflammatory cytokine (TNF-α, IL-1ß, and IL-6) production. CONCLUSIONS: In conclusion, the present study supports the effectiveness of BHBA in protecting dopaminergic neurons against inflammatory challenge.


Subject(s)
3-Hydroxybutyric Acid/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Inflammation/drug therapy , Inflammation/etiology , Parkinson Disease/complications , Receptors, G-Protein-Coupled/metabolism , Receptors, Nicotinic/metabolism , Animals , Calcium-Binding Proteins/metabolism , Cells, Cultured , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Embryo, Mammalian , Gene Expression Regulation/drug effects , Lipopolysaccharides/toxicity , Male , Mesencephalon/cytology , Microfilament Proteins/metabolism , Neuroglia/drug effects , Neurons/drug effects , Parkinson Disease/etiology , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Receptors, Nicotinic/genetics , Signal Transduction/drug effects , Stereotyped Behavior/drug effects
15.
Cell Physiol Biochem ; 34(2): 590-602, 2014.
Article in English | MEDLINE | ID: mdl-25116357

ABSTRACT

BACKGROUND/AIMS: GLP-2 has been shown to exert anti-inflammatory effects, but the underlying molecular mechanisms remained undefined. As macrophages are important in the development and maintenance of inflammation, we investigated whether exogenous GLP-2 modulates the expression of pro-inflammatory proteins in LPS stimulated murine peritoneal macrophages. METHODS: Macrophages were pretreated with various concentrations of GLP-2 for 1 h and then stimulated with LPS. The effects on pro-inflammatory enzymes (iNOS and COX-2), and pro-inflammatory cytokines (TNF-α, IL-1ß and IL-6) were analysed by Western blotting, ELISA and qRT-PCR. We also examined whether NF-κB or MAPK signaling was involved in the effects of GLP-2. RESULTS: In macrophages, GLP-2 blunted the effect of LPS on protein and mRNA expression levels of iNOS, COX-2, TNF-α, IL-1ß and IL-6. Pre-incubation of macrophages with GLP-2 also blunted LPS-induced IκB-α degradation, IκB-α phosphorylation and NF-κB translocation. In the presence of GLP-2, the effect of LPS treatment on ERK phosphorylation was also profoundly blunted. GLP-2 did, however, not significantly modify the effects of LPS on p38 and JNK activities. CONCLUSIONS: These findings demonstrate that in LPS primed macrophages, GLP-2 reduced pro-inflammatory enzymes and cytokine production via mechanisms involving the suppression of NF-κB activity and ERK phosphorylation.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Glucagon-Like Peptide 2/pharmacology , Inflammation/prevention & control , Lipopolysaccharides/antagonists & inhibitors , Macrophages, Peritoneal/drug effects , NF-kappa B/metabolism , Animals , Base Sequence , DNA Primers , Female , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred BALB C , Phosphorylation , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
16.
FEMS Microbiol Lett ; 351(1): 95-103, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24330077

ABSTRACT

Sortase A (SrtA), a transpeptidase, anchors surface proteins with an LPXTG-motif sorting signal to the cell envelope. To determine the role of SrtA in the pathogenesis of Staphylococcus aureus, we constructed a mutant strain, ∆SrtA, by genetic techniques and identified its functions in a S. aureus-induced mastitis mouse model. The histological and myeloperoxidase (MPO) level results showed that the ∆SrtA strain attenuated the inflammatory reaction in the mammary tissue of mice compared with wild-type S. aureus challenge. Additionally, the ELISA results showed that the ∆SrtA strain impaired the induction of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß) and interleukin-6 (IL-6), and the Western blot results showed that the mutant strain blocked the activation of nuclear factor-κB (NF-κB) and mitogen-activated protein kinases (MAPKs) by attenuating the degradation and phosphorylation of signaling pathway molecules such as IκBα, p65 and p38. These results suggest that SrtA is a key virulence factor in the pathogenesis of S. aureus-induced mastitis in mice. It appears that the srtA mutant affected the attachment of S. aureus to host cells, thus attenuating the activation of the NF-κB and MAPK signaling pathways, which regulated the expression of pro-inflammatory cytokines and decreased the susceptibility to mastitis.

17.
Mediators Inflamm ; 2013: 548073, 2013.
Article in English | MEDLINE | ID: mdl-24574581

ABSTRACT

The aim of this study was to investigate the anti-inflammatory effect of IL-21 on LPS-induced mouse peritoneal macrophages. The results showed that IL-21 significantly inhibited LPS-induced mRNA expression of IL-1ß, TNF-α, and IL-6 in macrophages, but not of IFN-γ, IL-10, CCL5, or CXCL2. ELISA analysis showed that IL-21 also suppressed LPS-induced production of TNF-α and IL-6 in culture supernatants. Western blot analysis showed that IL-21 clearly inhibited ERK and IκBα phosphorylation and NF-κB translocation in LPS-stimulated macrophages, but it increased STAT3 phosphorylation. Flow cytometric and Western blot analysis showed that IL-21 decreased M1 macrophages surface markers expression of CD86, iNOS, and TLR4 in LPS-stimulated cells. All results suggested that IL-21 decreases IL-6 and TNF-α production via inhibiting the phosphorylation of ERK and translocation of NF-κB and promotes a shift from the M1 to M2 macrophage phenotype by decreasing the expression of CD86, iNOS, and TLR4 and by increasing STAT3 phosphorylation in LPS-stimulated cells.


Subject(s)
Cytokines/metabolism , Interleukins/pharmacology , Macrophages, Peritoneal/cytology , Signal Transduction , Animals , B7-2 Antigen/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Flow Cytometry , Inflammation , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Lipopolysaccharides , Macrophages/cytology , Macrophages/metabolism , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred BALB C , Nitric Oxide Synthase Type II/metabolism , Phosphorylation , Real-Time Polymerase Chain Reaction , Recombinant Proteins/metabolism , STAT3 Transcription Factor/metabolism , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...