Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Int Med Res ; 48(10): 300060520964379, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33467974

ABSTRACT

OBJECTIVE: To observe the hemostatic effect of prophylactic uterine artery embolization (UAE) in patients with cesarean scar pregnancy (CSP) and to examine the risk factors for poor hemostasis. METHODS: Clinical data of 841 patients with CSP who underwent prophylactic UAE and curettage were retrospectively analyzed to evaluate the hemorrhage volume during curettage. A hemorrhage volume ≥200 mL was termed as poor hemostasis. The risk factors of poor hemostasis were analyzed and complications within 60 days postoperation were recorded. RESULTS: Among the 841 patients, 6.30% (53/841) had poor postoperative hemostasis. The independent risk factors of poor hemostasis were gestational sac size, parity, embolic agent diameter (>1000 µm), multivessel blood supply, and incomplete embolization. The main postoperative complications within 60 days after UAE were abdominal pain, low fever, nausea and vomiting, and buttock pain, with incidence rates of 71.22% (599/841), 47.44% (399/841), 39.12% (329/841), and 36.39% (306/841), respectively. CONCLUSIONS: Prophylactic UAE before curettage in patients with CSP is safe and effective in reducing intraoperative hemorrhage. Gestational sac size, parity, embolic agent diameter, multivessel blood supply, and incomplete embolization of all arteries supplying blood to the uterus are risk factors of poor hemostasis.


Subject(s)
Uterine Artery Embolization , Cesarean Section/adverse effects , Cicatrix/etiology , Cicatrix/prevention & control , Curettage , Female , Hemostasis , Humans , Pregnancy , Retrospective Studies , Risk Factors , Treatment Outcome
2.
Cell Mol Immunol ; 16(8): 694-705, 2019 08.
Article in English | MEDLINE | ID: mdl-29844591

ABSTRACT

Cystathionine-γ-lyase (CSE), an enzyme associated with hydrogen sulfide (H2S) production, is an important endogenous regulator of inflammation. Jumonji domain-containing protein 3 (JMJD3) is implicated in the immune response and inflammation. Here, we investigated the potential contribution of JMJD3 to endogenous CSE-mediated inflammation in rheumatoid arthritis (RA). Upregulated CSE and JMJD3 were identified in synovial fibroblasts (SFs) from RA patients as well as in the joints of arthritic mice. Knocking down CSE augmented inflammation in IL-1ß-induced SFs by increasing JMJD3 expression. In addition, CSE-/- mice with collagen-induced arthritis (CIA) developed severe joint inflammation and bone erosion. Conversely, overexpressing CSE inhibited JMJD3 expression by the transcription factor Sp-1 and was accompanied by reduced inflammation in IL-1ß-treated SFs. Furthermore, JMJD3 silencing or the administration of the JMJD3 inhibitor GSK-J4 significantly decreased the inflammatory response in IL-1ß-treated SFs, mainly by controlling the methylation status of H3K27me3 at the promoter of its target genes. GSK-J4 markedly attenuated the severity of arthritis in CIA mice. In conclusion, suppressing JMJD3 expression by the transcription factor Sp-1 is likely responsible for the ability of CSE to negatively modulate the inflammatory response and reduce the progression of RA.


Subject(s)
Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Autoimmunity/genetics , Cystathionine gamma-Lyase/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Autoimmunity/drug effects , Benzazepines/pharmacology , Benzazepines/therapeutic use , Cell Line , Cystathionine gamma-Lyase/genetics , Fibroblasts/metabolism , Gene Knockout Techniques , Humans , Inflammation/metabolism , Interleukin-1beta/pharmacology , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Jumonji Domain-Containing Histone Demethylases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Signal Transduction/drug effects , Signal Transduction/genetics , Synoviocytes/metabolism , Transfection
3.
Int J Nanomedicine ; 14: 10061-10077, 2019.
Article in English | MEDLINE | ID: mdl-31920303

ABSTRACT

PURPOSE: S-propargyl-cysteine (SPRC; alternatively known as ZYZ-802) is a novel modulator of endogenous tissue H2S concentrations with known cardioprotective and anti-inflammatory effects. However, its rapid metabolism and excretion have limited its clinical application. To overcome these issues, we have developed some novel liposomal carriers to deliver ZYZ-802 to cells and tissues and have characterized their physicochemical, morphological and pharmacological properties. METHODS: Two liposomal formulations of ZYZ-802 were prepared by thin-layer hydration and the morphological characteristics of each liposome system were assessed using a laser particle size analyzer and transmission electron microscopy. The entrapment efficiency and ZYZ-802 release profiles were determined following ultrafiltration centrifugation, dialysis tube and HPLC measurements. LC-MS/MS was used to evaluate the pharmacokinetic parameters and tissue distribution profiles of each formulation via the measurements of plasma and tissues ZYZ-802 and H2S concentrations. Using an in vivo model of heart failure (HF), the cardio-protective effects of liposomal carrier were determined by echocardiography, histopathology, Western blot and the assessment of antioxidant and myocardial fibrosis markers. RESULTS: Both liposomal formulations improved ZYZ-802 pharmacokinetics and optimized H2S concentrations in plasma and tissues. Liposomal ZYZ-802 showed enhanced cardioprotective effects in vivo. Importantly, liposomal ZYZ-802 could inhibit myocardial fibrosis via the inhibition of the TGF-ß1/Smad signaling pathway. CONCLUSION: The liposomal formulations of ZYZ-802 have enhanced pharmacokinetic and pharmacological properties in vivo. This work is the first report to describe the development of liposomal formulations to improve the sustained release of H2S within tissues.


Subject(s)
Cysteine/analogs & derivatives , Hydrogen Sulfide/therapeutic use , Myocardium/pathology , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Antioxidants/therapeutic use , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Cystathionine gamma-Lyase/metabolism , Cysteine/chemistry , Disease Models, Animal , Fibrosis , Heart Failure/drug therapy , Hydrogen Sulfide/blood , Liposomes , Male , Rats, Sprague-Dawley , Signal Transduction/drug effects
4.
Eur J Med Chem ; 160: 37-48, 2018 Dec 05.
Article in English | MEDLINE | ID: mdl-30317024

ABSTRACT

A series of novel pyridine-substituted piperidine derivatives were discovered as low nanomolar Ls-AChBP ligands with α7 nAChR partial agonism or antagonism activities. A high-resolution antagonist-bound Ls-AChBP complex was successfully resolved with a classic Loop C opening phenomenon and unique sulfur-π interactions which deviated from our previous docking mode to a large extent. With the knowledge of the co-complex, 27 novel piperidine derivatives were designed and synthesized. The structure-activity relationships (SARs) of the aromatic and pyridine regions were well established and binding modes were illustrated with the help of molecular docking which indicated that interactions with Trp 143 and the "water bridge" are essential for the high binding affinities. Halogen bonding as well as the space around 5'- or 6'- position of the pyridine ring was also proposed to influence the binding conformation of the compounds. Notably, two enantiomers of compound 2 showed opposite functions toward α7 nAChR and compound (S)-2 showed sub-nanomolar affinity (Ki = 0.86 nM) on Ls-AChBP and partial agonism (pEC50 = 4.69 ±â€¯0.11,Emax = 36.1%) on α7 nAChR with reasonable pharmacokinetics (PK) properties and fine ability of blood-brain-barrier (BBB) penetration. This study provided promising hits to develop candidates targeting nAChR-related CNS diseases.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Drug Discovery , Nicotinic Agonists/pharmacology , Piperidines/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/agonists , Animals , Carrier Proteins/metabolism , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Ligands , Lymnaea , Models, Molecular , Molecular Structure , Nicotinic Agonists/chemical synthesis , Nicotinic Agonists/chemistry , Piperidines/chemical synthesis , Piperidines/chemistry , Structure-Activity Relationship , alpha7 Nicotinic Acetylcholine Receptor/metabolism
5.
Cell Physiol Biochem ; 47(2): 458-474, 2018.
Article in English | MEDLINE | ID: mdl-29794432

ABSTRACT

BACKGROUND/AIMS: Sepsis is a severe and complicated syndrome that is characterized by dysregulation of host inflammatory responses and organ failure. Cystathionine-γ-lyase (CSE)/ hydrogen sulfide (H2S) has potential anti-inflammatory activities in a variety of inflammatory diseases. NADPH oxidase 4 (Nox4), a member of the NADPH oxidases, is the major source of reactive oxygen species (ROS) and its expression is increased in sepsis, but its function in CSE-mediated anti-inflammatory activities remains unknown. METHODS: Macrophages were either transfected with CSE, Nox4 siRNA or transduced with lentiviral vector encoding CSE or Nox4, and then stimulated with lipopolysaccharide (LPS). The expression of inflammatory mediators and signaling pathway activation were measured by quantitative PCR (qPCR), ELISA, and immunoblotting. LPS-induced shock severity in WT, Nox4 knockdown and CSE knockout (CSE-/-) mice was assessed. RESULTS: Here we showed that CSE and Nox4 were upregulated in macrophage and mouse in response to LPS. After LPS stimulation, the inflammatory responses were significantly ameliorated by lentiviral Nox4 shRNA knockdown, but were exacerbated by lentiviral overexpressing Nox4. Furthermore, Nox4 mediated inflammation through PI3K/Akt and p-p38 mitogen-activated protein kinase signal pathway. Notably, CSE knockout served to amplify the inflammatory cascade by increasing Nox4-ROS signaling activation in septic mice and macrophage. Similarly, the enhanced production of inflammatory mediators by macrophages was reduced by CSE overexpression. CONCLUSION: Thus, we demonstrated that CSE/H2S attenuated LPS-induced sepsis against oxidative stress and inflammation damage probably largely through mediated Nox4 pathway.


Subject(s)
Hydrogen Sulfide/pharmacology , Lipopolysaccharides/pharmacology , NADPH Oxidase 4/metabolism , Oxidative Stress/drug effects , Animals , Cystathionine gamma-Lyase/deficiency , Cystathionine gamma-Lyase/genetics , Cytokines/analysis , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 4/antagonists & inhibitors , NADPH Oxidase 4/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RAW 264.7 Cells , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Cell Death Dis ; 9(5): 503, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29717129

ABSTRACT

Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by abnormal inflammation, angiogenesis, and cartilage destruction. In RA, neoangiogenesis is an early and crucial event to promote the formation of pannus, causing further inflammatory cell infiltration. The transcription factor GATA4 is a critical regulator of cardiac differentiation-specific gene expression. We find that a higher level of GATA4 exists in synovium of rheumatoid arthritis (RA) patients, but the function of GATA4 in RA remains unclear. In the present study, IL-1ß induces inflammation in fibroblast-like synoviocytes (FLS) MH7A, which is accompanied with the increased expression of GATA4 and VEGF production. Through application of GATA4 loss-of-function assays, we confirm the requirement of GATA4 expression for inflammation induced by IL-1ß in FLS. In addition, we demonstrate for the first time that GATA4 plays key roles in regulating VEGF secretion from RA FLS to promote cellular proliferation, induce cell migration, and angiogenic tube formation of endothelial cells. GATA4 induces the angiogenic factors VEGFA and VEGFC, by directly binding to the promoter and enhancing transcription. The knockdown of GATA4 attenuates the development of collagen-induced arthritis (CIA) and prevents RA-augmented angiogenesis in vivo, which are accompanied with decreased VEGF level. These results reveal a previously unrecognized function for GATA4 as a regulator of RA angiogenesis and we provide experimental data validating the therapeutic target of GATA4 in RA mice.


Subject(s)
Arthritis, Experimental/genetics , Arthritis, Rheumatoid/genetics , GATA4 Transcription Factor/genetics , Neovascularization, Pathologic/genetics , Synoviocytes/metabolism , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Arthritis, Experimental/prevention & control , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , GATA4 Transcription Factor/antagonists & inhibitors , GATA4 Transcription Factor/metabolism , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Male , Mice, Inbred DBA , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/prevention & control , Promoter Regions, Genetic , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Synovial Membrane/metabolism , Synovial Membrane/pathology , Synoviocytes/drug effects , Synoviocytes/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/metabolism
7.
Cardiovasc Res ; 114(7): 1016-1028, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29529137

ABSTRACT

Aims: Angiotensin II (Ang II) causes vascular inflammation, leading to vascular endothelial cell dysfunction, and is associated with the development of cardiovascular diseases. Therefore, interventions in inflammation may contribute to the reduction of cardiovascular diseases. Here, we aim to demonstrate that HDAC4, one of class IIa family histone de-acetylases (HDACs) members, promotes autophagy-dependent vascular inflammation. Methods and results: By loss-of-function approaches, our study provides the first evidence that HDAC4 mediates Ang II-induced vascular inflammation in vitro and in vivo. In response to the Ang II, HDAC4 expression is up-regulated rapidly, with increased autophagic flux and inflammatory mediators in vascular endothelial cells (VECs). In turn, HDAC4 deficiency suppresses activation of autophagy, leading to reduced inflammation in Ang II-induced VECs. Consistently, using autophagy inhibitor or silencing LC3-II also alleviates vascular inflammation. Furthermore, HDAC4 regulates autophagy via facilitating transcription factor forkhead box O3a (FoxO3a) de-acetylation, thereby to increase its transcriptional activity. Loss of HDAC4 in VECs results in inhibition of FoxO3a de-acetylation to block its transcriptional activity, leading to downregulation of the downstream FoxO3a target, and hence reduces autophagy and vascular inflammation. FoxO3a silencing using siRNA approach significantly inhibits activation of autophagy. Finally, knockdown of HDAC4 in Ang II-infused mouse models ameliorates vascular inflammation, suggesting that inhibitor of HDAC4 may be potential therapeutics for vascular diseases associated with inflammation. Conclusion: These results suggest that HDAC4-mediated FoxO3a acetylation regulates Ang II-induced autophagy activation, which in turn plays an essential role in causing vascular inflammation.


Subject(s)
Autophagy , Histone Deacetylases/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Vasculitis/enzymology , Acetylation , Angiotensin II , Animals , Cells, Cultured , Disease Models, Animal , Forkhead Box Protein O3/genetics , Forkhead Box Protein O3/metabolism , Histone Deacetylases/genetics , Male , Mice, Inbred C57BL , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Protein Processing, Post-Translational , Rats , Rats, Sprague-Dawley , Signal Transduction , Vasculitis/chemically induced , Vasculitis/genetics , Vasculitis/pathology
8.
FASEB J ; 32(7): 4031-4042, 2018 07.
Article in English | MEDLINE | ID: mdl-29481307

ABSTRACT

Rheumatoid arthritis (RA) is an immune-mediated disease with the characteristics of progressive joint destruction, deformity, and disability. Epigenetic changes have been implicated in the development of some autoimmune disorders, resulting in an alteration of gene transcription. Here, we investigated how Jumonji C family of histone demethylases (JMJD3) regulated the proliferation and activation of fibroblast-like synoviocytes (FLSs), which are involved in RA joint destruction and pathologic process. The JMJD3 expression and proliferation markers in RA-FLS were higher than those in healthy-FLS and were upregulated in platelet-derived growth factor (PDGF)-induced FLS. Elevated JMJD3 promoted the proliferation and migration of FLS. Treatment with JMJD3 small interfering RNA or inhibitor glycogen synthase kinase (GSK) J4 led to decreased proliferation and migration of FLS. Interestingly, induction of proliferating cell nuclear antigen (PCNA), a major player of the cell-cycle regulation, was correlated with trimethylated lysine 27 in histone H3 loss around the gene promoters. The knockdown of JMJD3 abolished PCNA expression in PDGF-induced FLS and further inhibited cell proliferation and migration, suggesting that JMJD3/PCNA played a crucial role in aspects of FLS proliferation and migration. In vivo, the ability of GSK J4 to hinder collagen-induced arthritis (CIA) in DBA/1 mice was evaluated. We found that GSK J4 markedly attenuated the severity of arthritis in CIA mice. The therapeutic effects were associated with ameliorated joint swelling and reduced bone erosion and destruction. This study revealed how JMJD3 integrated with epigenetic processes to regulate RA-FLS proliferation and invasion. These data suggested that JMJD3 might contribute to rheumatoid synovial hyperplasia and have the potential as a novel therapeutic target for RA.-Jia, W., Wu, W., Yang, D., Xiao, C., Su, Z., Huang, Z., Li, Z., Qin, M., Huang, M., Liu, S., Long, F., Mao, J., Liu, X., Zhu, Y. Z. Histone demethylase JMJD3 regulates fibroblast-like synoviocyte-mediated proliferation and joint destruction in rheumatoid arthritis.


Subject(s)
Arthritis, Rheumatoid/metabolism , Cell Proliferation , Jumonji Domain-Containing Histone Demethylases/metabolism , Synoviocytes/metabolism , Animals , Antirheumatic Agents/pharmacology , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Benzazepines/pharmacology , Benzazepines/therapeutic use , Cell Movement , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/physiology , Joints/drug effects , Joints/metabolism , Joints/pathology , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Jumonji Domain-Containing Histone Demethylases/genetics , Male , Mice , Mice, Inbred DBA , Proliferating Cell Nuclear Antigen/metabolism , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Rats , Rats, Sprague-Dawley , Synoviocytes/drug effects , Synoviocytes/physiology
9.
Biochem Pharmacol ; 149: 153-162, 2018 03.
Article in English | MEDLINE | ID: mdl-29074105

ABSTRACT

Overproduction of inflammatory mediators contributes to uncontrolled inflammation during endotoxin shock. Cystathionine-γ-lyase (CSE), an enzyme involved in hydrogen sulfide (H2S) biosynthesis, has potential anti-inflammatory activity in a variety of inflammatory diseases. Jumonji domain-containing protein 3 (JMJD3), a histone 3 Lys27 (H3K27) demethylase, has been implicated in macrophage activation, but its function in CSE-mediated anti-inflammatory activities remains unknown. In the present study CSE was found to be upregulated in macrophages and mouse lipopolysaccharide (LPS) challenge models. LPS stimulation also enhanced the activation of JMJD3 and decreased H3K27me3 levels. JMJD3 knockdown upregulated H3K27me3 levels and attenuated the LPS-mediated inflammatory response. CSE knockout amplified the inflammatory cascade by increasing JMJD3 expression in septic mice. Similarly, enhanced production of inflammatory mediators by macrophages was mitigated by CSE overexpression via inhibition of JMJD3 expression. This is the first report indicating that inflammation enhanced CSE/H2S system biosynthesis, that in turn attenuated the LPS-triggered inflammatory response by regulating JMJD3 expression. Thus, the CSE/H2S system represents an epigenetic-based modification mechanism to prevent uncontrolled inflammation.


Subject(s)
Inflammation/chemically induced , Jumonji Domain-Containing Histone Demethylases/metabolism , Lipopolysaccharides/toxicity , Shock, Septic/chemically induced , Animals , Cystathionine gamma-Lyase/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Gene Knockdown Techniques , Histones/genetics , Histones/metabolism , Hydrogen Sulfide , Inflammation/metabolism , Macrophages , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Shock, Septic/metabolism
10.
Phytomedicine ; 32: 15-23, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28732803

ABSTRACT

BACKGROUND: Endothelial inflammation is an increasingly prevalent condition in the pathogenesis of many cardiovascular diseases. (-)-7(S)-hydroxymatairesinol (7-HMR), a naturally occurring plant lignan, possesses both antioxidant and anti-cancer properties and therefore would be a good strategy to suppress tumor necrosis factor-α (TNF-α)-mediated inflammation in vascular endothelial cells (VECs). PURPOSE: The objective of this study is to evaluate for its anti-inflammatory effect on TNF-α-stimulated VECs and underling mechanisms. STUDY DESIGN/METHODS: The effect of the 7-HMR on suppression of TNF-α-induced inflammation mediators in VECs were determined by qRT-PCR and Western blot. MAPKs and phosphorylation of Akt, HO-1 and NF-κB p65 were examined using Western blot. Nuclear localisation of NF-κB was also examined using Western blot and immunofluorescence. RESULTS: Here we found that 7-HMR could suppress TNF-α-induced inflammatory mediators, such as vascularcelladhesion molecule-1, interleukin-6 and inducible nitric oxide synthase expression both in mRNA and protein levels, and concentration-dependently attenuated reactive oxidase species generation. We further identified that 7-HMR remarkably induced superoxide dismutase and heme oxygenase-1 expression associated with degradation of Kelch-like ECH-associated protein 1 (keap1) and up-regulated nuclear factor erythroid 2-related factor 2 (Nrf2). In addition, 7-HMR time- and concentration-dependently attenuated TNF-α-induced phosphorylation of extracellular signal-regulated kinase 1/2 (ERK) and Akt, but not p38, or c-Jun N-terminal kinase 1/2. Moreover, 7-HMR significantly suppressed TNF-α-mediated nuclear factor-κB (NF-κB) activation by inhibiting phosphorylation and nuclear translocation of NF-κB p65. CONCLUSION: Our results demonstrated that 7-HMR inhibited TNF-α-stimulated endothelial inflammation, at least in part, through inhibition of NF-κB activation and upregulation of Nrf2-antioxidant response element signaling pathway, suggesting 7-HMR might be used as a promising vascular protective drug.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Inflammation/drug therapy , Lignans/pharmacology , NF-kappa B/metabolism , Animals , Antioxidant Response Elements/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Heme Oxygenase-1/metabolism , Inflammation/metabolism , Inflammation Mediators/metabolism , Interleukin-6/metabolism , Male , NF-E2-Related Factor 2/metabolism , Phosphorylation/drug effects , Rats , Tumor Necrosis Factor-alpha/adverse effects , Tumor Necrosis Factor-alpha/metabolism
11.
Neurochem Res ; 42(10): 2850-2860, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28512713

ABSTRACT

Chronic neuroinflammation is a pathological feature of neurodegenerative diseases. Inhibition of microglia-mediated neuroinflammation might be a potential strategy for neurodegeneration. Matairesinol, a dibenzylbutyrolactone plant lignan, presents in a wide variety of foodstuffs. It has been found to possess anti-angiogenic, anti-oxidative, anti-cancer and anti-fungal activities. In the present study, we investigated the anti-neuroinflammation effects of matairesinol on lipopolysaccharide (LPS)-induced BV2 microglia cells and the related molecular mechanisms. The results showed that matairesinol inhibited microglia activation by reducing the production of nitric oxide, the expression of inducible nitric oxide synthase and cyclooxygenase-2 in a concentration-dependent manner (6.25, 12.5, 25 µM). In the molecular signaling pathway, LPS-induced nuclear factor-kappa B (NF-κB) transcriptional activity and translocation into the nucleus were remarkably suppressed by matairesinol through the inhibition of the extracellular signal-regulated kinase (ERK)1/2 signal transduction pathways, but not p38 MAPK or c-jun N-terminal kinase (JNK). Meanwhile, matairesinol also blocked LPS-mediated microglia migration and this was associated with inhibition of LPS-induced Src phosphorylation as well as Src expression in a concentration-dependent manner. Taken together, these results suggest that matairesinol inhibited inflammatory response and migration in LPS-induced BV2 microglia, and the mechanisms may be associated with the NF-κB activation and modulation of Src pathway.


Subject(s)
Furans/pharmacology , Lignans/pharmacology , MAP Kinase Signaling System/drug effects , Microglia/drug effects , NF-kappa B/metabolism , Signal Transduction/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Lipopolysaccharides/pharmacology , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Oncogene Protein pp60(v-src)/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
12.
Redox Biol ; 10: 157-167, 2016 12.
Article in English | MEDLINE | ID: mdl-27744121

ABSTRACT

Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disorder. Hydrogen sulfide (H2S), the third physiological gasotransmitter, is well recognized as an anti-inflammatory mediator in various inflammatory conditions. Herein, we explored the protective effects of S-propargyl-cysteine (SPRC, also known as ZYZ-802), an endogenous H2S modulator, on RA and determined the underlying mechanisms. In the present study, SPRC concentration-dependently attenuated inflammatory mediator expression, reactive oxidase species generation, and the expression and activity of matrix metalloproteinases (MMP)-9 in interleukin (IL)-1ß-induced human rheumatoid fibroblast-like synoviocytes MH7A. In addition, SPRC blocked IL-1ß-mediated migration and invasion of MH7A cells. As expected, the protective effects of SPRC were partially abrogated by DL-propargylglycine (PAG, a H2S biosynthesis inhibitor). In vivo study also demonstrated that SPRC treatment markedly ameliorated the severity of RA in adjuvant-induced arthritis rats, and this effect was associated with the inhibition of inflammatory response. We further identified that SPRC remarkably induced heme oxygenase-1 expression associated with the degradation of Kelch-like ECH-associated protein 1 (Keap1) and nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2); this effect was attributed to the sulfhydrylation of the cysteine residue of Keap1. Our data demonstrated for the first time that SPRC, an endogenous H2S modulator, exerted anti-inflammatory properties in RA by upregulating the Nrf2-antioxidant response element (ARE) signaling pathway.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Antioxidant Response Elements/drug effects , Arthritis, Rheumatoid/drug therapy , Cysteine/analogs & derivatives , NF-E2-Related Factor 2/genetics , Animals , Anti-Inflammatory Agents/pharmacology , Arthritis, Rheumatoid/genetics , Cell Line , Cell Movement/drug effects , Cell Survival , Cysteine/administration & dosage , Cysteine/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Male , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Up-Regulation
13.
Nat Prod Commun ; 11(11): 1757-1761, 2016 Nov.
Article in English | MEDLINE | ID: mdl-30475522

ABSTRACT

Herba Leonuri, also named Chinese Motherwort, has been extensively investigated as an effective agent on the uterus system. Our group has been studying the natural products of Herba Leonuri for several years, and during this period, many biological activities of the drug were recognized. Leonurine (4-guanidino-N- butyl-syringate) is an alkaloid present in Herba Leonuri. Recently, growing evidence has highlighted the therapeutic potential of leonurine in multiple diseases, especially cardiovascular. In this review, we discuss the biological activities of leonurine, as well as recent advances involving this alkaloid.


Subject(s)
Gallic Acid/analogs & derivatives , Leonurus/chemistry , Medicine, Chinese Traditional , Gallic Acid/chemistry , Gallic Acid/pharmacology , Humans , Molecular Structure , Phytotherapy
SELECTION OF CITATIONS
SEARCH DETAIL
...