Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Immunopharmacol Immunotoxicol ; : 1-11, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38772618

ABSTRACT

BACKGROUND: Systemic inflammatory response syndrome (SIRS) is an uncontrolled systemic inflammatory response. Proanthocyanidins (PC) is a general term of polyphenol compounds widely existed in blueberry fruits and can treat inflammation-related diseases. This study aimed to explore the regulatory effect of PC on lipopolysaccharide (LPS)-induced systemic inflammation and its potential mechanism, providing effective strategies for the further development of PC. METHODS: Here, RAW264.7 macrophages were stimulated with LPS to establish an inflammation model in vitro, while endotoxin shock mouse models were constructed by LPS in vivo. The function of PC was investigated by MTT, ELISA kits, H&E staining, immunohistochemistry, and Western blot analysis. RESULTS: Functionally, PC could demonstrate the potential to mitigate mortality in mice with endotoxin shock, as well as attenuated the levels of inflammatory cytokines (IL-6, TNF-α) and biochemical indicators (AST, ALT, CRE and BUN). Moreover, it had a significant protective effect on lung and kidney tissues damage. Mechanistically, PC exerted anti-inflammatory effects by inhibiting the activation of the NF-κB/NLRP3 signaling pathway. CONCLUSION: PC might have the potential ability of anti-inflammatory effects via modulation of the NF-κB/NLRP3 signaling pathway.

2.
Int Immunopharmacol ; 129: 111538, 2024 Mar 10.
Article in English | MEDLINE | ID: mdl-38306830

ABSTRACT

CCl4-induced acute liver injury (ALI) is characterized by heightened autophagy, inflammation, and oxidative damage. Accumulating evidence suggests that harmine exerts beneficial effects in countering CCl4-induced ALI by mitigating inflammation and oxidative stress. However, the impact of autophagy on CCl4-induced ALI and the protective role of harmine remain unclear. This study aimed to investigate the potential protective effects of harmine against CCl4-induced ALI in mice by suppressing autophagy and inflammation. Male Kunming mice were orally administered harmine or bifendate for seven days. Subsequently, one hour after the final administration, the model group and treatment groups were intraperitoneally injected with CCl4 to induce ALI. The findings revealed that harmine significantly reduced the levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT) in serum, and ameliorated the liver histopathological changes induced by CCl4. Furthermore, harmine diminished the levels of TNF-α and IL-6, restored the levels of glutathione (GSH) and superoxide dismutase (SOD), and suppressed the production of nitric oxide (NO) and malondialdehyde (MDA) in the liver. Mechanistically, harmine down-regulated LC3B II/I, p38 MAPK, TLR4, and NF-κB levels, while upregulating p62, Bcl-2, Beclin1, ULK1, and p-mTOR expression. In conclusion, harmine mitigated CCl4-induced ALI by inhibiting autophagy and inflammation through the p38 MAPK/mTOR autophagy pathway, the Bcl-2/Beclin1 pathway, and the TLR4/NF-κB pathway.


Subject(s)
Harmine , NF-kappa B , Mice , Male , Animals , NF-kappa B/metabolism , Harmine/pharmacology , Harmine/therapeutic use , Toll-Like Receptor 4/metabolism , Beclin-1/metabolism , Liver/pathology , Inflammation/metabolism , Glutathione/metabolism , TOR Serine-Threonine Kinases/metabolism , Autophagy , Proto-Oncogene Proteins c-bcl-2/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Reprod Biol ; 23(3): 100789, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37499346

ABSTRACT

ß-endorphin (ß-EP) is involved in the regulation of male germ cells; however, little is known about the effect of ß-EP on primary germ cells via opioid receptors. In this study, we first revealed significant cell apoptosis in the testis of male rats after ß-EP intervention. Subsequently, the expression of the mu opioid receptor (MOR) was detected in both Leydig cells (LCs) and spermatogonia (SGs) by fluorescence colocalization; overlapping signals were also detected in apoptotic cells. In addition, LCs and SGs were separated from the testis of male rats and primary cells were treated with ß-EP; this increased the mRNA levels of MOR and was accompanied by acute cell apoptosis. Our findings provide a foundation for the further study of apoptosis in reproductive cells regulated by ß-EP and the MOR receptor.


Subject(s)
Testis , beta-Endorphin , Rats , Animals , Male , Testis/metabolism , beta-Endorphin/genetics , beta-Endorphin/metabolism , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Leydig Cells/metabolism , Apoptosis
4.
Immunopharmacol Immunotoxicol ; 44(5): 641-655, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35506641

ABSTRACT

BACKGROUND: Tectoridin, widely extracted and separated from the rhizome of Iris tectorum Maxim, is extensively reported to have affluent bioactivity, but rarely reported to have anti-inflammatory effects. In this study, we aim to investigate the anti-inflammatory effects and the underlying mechanisms of tectoridin. METHODS: Here, RAW264.7 macrophages were stimulated with Lipopolysaccharide (LPS) for the inflammation model in vitro. Experimental animals received tectoridin and Dexamethasone (DEX) before LPS injection for endotoxic shock mouse model in vivo. The pro-inflammatory mediators and cytokines in the cell supernatant and serum were detected by ELISA kits. The tissue damages were assessed by biochemical indexes and H&E staining. Immunohistochemistry and Western blot were performed for the detection of proteins. RESULTS: Our data showed that tectoridin attenuated the LPS-up-regulated nitric oxide (NO), interleukin-6 (IL-6), and interleukin-18 (IL-18) from macrophages and tumor necrosis factor-α (TNF-α); (IL-6) and (IL-1ß) in the serum levels. Besides, our histopathological study showed that the damages caused by LPS in the lung, liver, and kidney tissues were decreased. Furthermore, our results demonstrated that tectoridin inhibited the activation of TLR4-NF-κB/NLRP3 signaling proved by immunohistochemistry assay and Western blot. CONCLUSION: Taken all together, tectoridin might have the potential ability of anti-inflammatory effects and the possible mechanism may be relevant to its inhibition of TLR4-NF-κB/NLRP3 signaling.


Subject(s)
Lipopolysaccharides , NF-kappa B , Animals , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Dexamethasone/pharmacology , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Inflammation Mediators/metabolism , Interleukin-18 , Interleukin-6 , Isoflavones , Lipopolysaccharides/toxicity , Mice , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nitric Oxide/metabolism , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha
5.
Eur J Pharmacol ; 891: 173761, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33249078

ABSTRACT

Vascular smooth muscle cells (VSMCs) lesions play an important role in atherosclerosis. The latest findings indicate that green tea extract has potential benefits for patients with atherosclerosis, but the components and mechanisms of action are unknown. (-)-Epicatechin gallate (ECG) is the main active ingredient extracted from green tea and has significant biological functions. However, the mechanism of ECG in atherosclerosis remains unclear. Therefore, we investigated the intervention of ECG on VSMCs induced by oxidized low-density lipoprotein (ox-LDL). The results show that ECG reduces the inflammatory response by preventing the overproduction of inflammatory mediators in VSMCs. ECG regulates the cell cycle and down-regulates the expression of proliferating cell nuclear antigen (PCNA) and cyclinD1, and then exerts an anti-proliferative effect. Furthermore, inhibition of the expression of matrix metalloproteinase 2 (MMP-2) and intercellular adhesion molecule 1 (ICAM-1) may be the mechanism by which ECG inhibits the migration of ox-LDL-induced VSMCs. Oil red O staining results show that ECG can improve cell foaming and reduce the content of total cholesterol (TC). In addition, ECG significantly reduces reactive oxygen species activity and also reduces the expression of p-p38, p-JNK, p-ERK1/2, p-IκBα, p-NF-κBp65, and TLR4. These results indicate that ECG has potential clinical applications for preventing atherosclerosis.


Subject(s)
Atherosclerosis/drug therapy , Catechin/analogs & derivatives , Cell Movement/drug effects , Cell Proliferation/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Catechin/pharmacology , Cells, Cultured , Foam Cells/drug effects , Foam Cells/metabolism , Foam Cells/pathology , Inflammation Mediators/metabolism , Intercellular Adhesion Molecule-1/metabolism , Lipoproteins, LDL/pharmacology , Matrix Metalloproteinase 2/metabolism , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Phosphorylation , Plaque, Atherosclerotic , Rats , Reactive Oxygen Species/metabolism , Signal Transduction
6.
Int Immunopharmacol ; 90: 107170, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33218940

ABSTRACT

Inflammation plays an important role in the process of atherosclerosis (AS). Inhibition of inflammation is an effective way to prevent AS. Imperatorin (IMP) is a kind of furan coumarin with various activities. In this study, the anti-inflammatory effect of IMP was explored in oxidized low-density lipoprotein (ox-LDL)-induced VSMCs and high fat diet (HFD)-induced ApoE-/- mice. The results showed that IMP attenuated the elevation of TNF-α, IL-6, MCP-1 and NO induced by ox-LDL in supernatant of VSMCs. IMP has normalized the levels of serum lipids (TC, TG, LDL-C and HDL-C) and attenuated inflammatory cytokines in serum. IMP also improved pathological changes and lipid accumulation in aorta. Matrix metalloproteinase-2 (MMP-2) expression in aorta was down-regulated by IMP. IMP could inhibit the phosphorylation of MAPKs pathway in the aorta and VSMCs, resulting in a significant decrease in the contents of p-ERK 1/2, p-JNK and p-P38. Overall, IMP could exert anti-inflammatory effects in vivo and in vitro to interfere with AS.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Atherosclerosis/metabolism , Furocoumarins/pharmacology , Inflammation/drug therapy , MAP Kinase Signaling System/drug effects , Animals , Aorta/drug effects , Aorta/physiopathology , Cytokines/blood , Cytokines/drug effects , Cytokines/metabolism , Lipid Metabolism/drug effects , Lipids/blood , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal
7.
Phytother Res ; 35(4): 2220-2229, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33350533

ABSTRACT

Atherosclerosis (AS) is a common chronic inflammatory disease of the arteries, which is closely related to dyslipidemia, inflammatory factors, and oxidative stress. Poria cocos polysaccharides (PCP) are one of the main active ingredients of Poria, which has significant pharmacological effects. In this study, the potential protective mechanism of PCP on AS was discussed in the ApoE-/- mice model induced by high-fat diet. These pathological changes were evaluated by H&E and oil red O staining. The levels of pro-inflammatory cytokines in aortic tissue were measured by enzyme-linked immunosorbent assay kit. These protein expressions were detected by Western blot and immunohistochemistry. The results showed that PCP inhibited the serum inflammatory mediators (tumor necrosis factor-α, interleukin-6, and nitric oxide) and lipids (low-density lipoprotein-cholesterol, triglyceride, and total cholesterol) increase. Moreover, PCP also reduced the concentration of malondialdehyde, increased the activity of superoxide dismutase, and improved the pathological changes of the aorta. Finally, PCP inhibited the activation of the TLR4/NF-κB pathway in the aorta and blocked the expression of matrix metalloproteinase 2 and intercellular adhesion molecule 1 proteins. In short, PCP intervenes in AS by reducing inflammatory factors and blood lipid levels.


Subject(s)
Apolipoproteins E/metabolism , Atherosclerosis/drug therapy , Diet, High-Fat/adverse effects , Inflammation/drug therapy , Wolfiporia/chemistry , Animals , Disease Models, Animal , Male , Mice , Signal Transduction
8.
Int Immunopharmacol ; 80: 106173, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31945610

ABSTRACT

Oxidative stress, inflammation, and foam cell formation in vascular smooth muscle cells (VSMCs) are considered to play crucial roles in the pathogenesis of atherosclerosis. Poria cocos polysaccharides (PCP) has been shown to possess anti-inflammatory, antitumor and anti-oxidative properties. In this study we explored the effects of PCP on ox-LDL-induced inflammation, oxidative stress and foam cell formation in VSMCs. PCP significantly attenuated ox-LDL-induced oxidative stress, as evidenced by the decreased reactive oxygen species (ROS) and MDA levels, and the increased SOD activity in VSMCs. PCP suppressed the induction effect of ox-LDL on inflammatory cytokines and inflammatory mediators. PCP also substantially inhibited VSMCs foam cell formation and intracellular lipids accumulation. Mechanistically, PCP suppressed ox-LDL-induced up-regulation of LOX-1, which is responsible for ox-LDL uptake. Western blotting suggested that PCP activated ERK1/2 signaling pathway, increased Nrf2 translocated from cytoplasm to nucleus and heme oxygenase-1 (HO-1) expression. Up-regulation of PCP on Nrf2/HO-1 signaling was reversed by pretreatment with ERK inhibitor PD98059, indicating the involvement of ERK in PCP activation of Nrf2/HO-1 signaling. In conclusion, these results demonstrated that PCP exerted its protection against oxidative stress and inflammation via the ERK/Nrf2/HO-1 signaling pathway and that PCP may be a promising candidate for the therapy of atherosclerosis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Atherosclerosis/drug therapy , Fungal Polysaccharides/pharmacology , Myocytes, Smooth Muscle/drug effects , Signal Transduction/drug effects , Wolfiporia/chemistry , Atherosclerosis/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Foam Cells/drug effects , Foam Cells/immunology , Heme Oxygenase-1/metabolism , Humans , Lipid Metabolism/drug effects , Lipid Metabolism/immunology , Lipoproteins, LDL/immunology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Oxidative Stress/immunology , Reactive Oxygen Species/metabolism , Signal Transduction/immunology
9.
Fundam Clin Pharmacol ; 34(1): 91-101, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31325387

ABSTRACT

Fraxin, the effective component isolated from Cortex Fraxini, has been reported to have anti-inflammation effects. The aim of this study was to explore the effect of fraxin on lipopolysaccharide (LPS)-induced endotoxic shock in mice. We used Kunming male mice to establish the model, and we found that fraxin could improve the survival rate of the LPS-induced mice. Histopathological study showed that fraxin could mitigate the injuries in LPS-induced lung and liver tissues. The levels of tumour necrosis factor-α and interleukin-6 both in serum and lung, liver tissues, and the productions of nitric oxide (NO), aspartate transaminase and alanine transaminase in serum were decreased by fraxin. Western blot assay demonstrated that the pretreatment with fraxin could downregulate LPS-induced protein expressions of nuclear factor-kappa B (NF-κB) and NLRP3 inflammatory corpuscle signalling pathways. Overall, fraxin had protective effects on LPS-induced endotoxic shock mice and the possible mechanisms might activate through NF-κB and NLRP3 inflammatory corpuscle signalling pathways.


Subject(s)
Coumarins/pharmacology , Cytokines/metabolism , Inflammation/drug therapy , Shock, Septic/prevention & control , Animals , Anti-Inflammatory Agents/pharmacology , Disease Models, Animal , Inflammation/pathology , Lipopolysaccharides/toxicity , Male , Mice , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nitric Oxide/metabolism , Signal Transduction/drug effects , Survival Rate
10.
Int Immunopharmacol ; 81: 106024, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31784404

ABSTRACT

Acute lung injury (ALI) is a common lung disease accompanied by acute and persistent pulmonary inflammatory response syndrome, which leads to alveolar epithelial cells and capillary endothelial cell damage. Yam glycoprotein, separated from traditional Chinese yam, has been shown to have anti-inflammatory and immunomodulatory effects. In this experiment, we mainly studied the therapeutic effect and mechanism of a glycoprotein on the lipopolysaccharide (LPS)-induced ALI mice. An oral glycoprotein method was used to treat the mouse ALI model induced by LPS injection in the peritoneal cavity. Afterward, we measured the wet/dry (W/D) ratio, the activity of myeloperoxidase (MPO), the oxidative index superoxide dismutase (SOD), malondialdehyde (MDA), glutathione peroxidase (GSH-PX) and the production of inflammatory cytokines interleukin-1ß (IL-1ß), tumour necrosis factor-α (TNF-α), and interleukin-6 (IL-6) to evaluate the effect of yam glycoprotein on lung tissue changes. We examined the protein expression of TLR4, ASC, NF-κBp65, p-NF-κBp65, Caspase-1, IκB, NLRP3, p-IκB, and ß-actin by western blot analysis. Immunohistochemical analyses of NLRP3 and p-p65 in lung tissue were carried out to assess the mechanism of glycoprotein action. This result suggests that glycoprotein markedly depressed LPS-induced lung W/D ratio, MPO activity, MDA content SOD and GSH-Px depletion, and the contents of inflammatory cytokines IL-1ß, IL-6, and TNF-α. Moreover, glycoprotein blocked TLR4/NF-κBp65 signaling activation and NLRP3inflammasome expression in LPS-induced ALI mice. As this particular study shows, glycoprotein has a safeguarding effects on LPS-induced ALI mice, possibly via activating NLRP3inflammasome and TLR4/NF-κB signaling pathways.


Subject(s)
Acute Lung Injury/drug therapy , Anti-Inflammatory Agents/pharmacology , Dioscorea , Glycoproteins/pharmacology , Plant Proteins/pharmacology , Signal Transduction/drug effects , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Animals , Anti-Inflammatory Agents/therapeutic use , Disease Models, Animal , Glycoproteins/isolation & purification , Glycoproteins/therapeutic use , Humans , Inflammasomes/drug effects , Inflammasomes/immunology , Inflammasomes/metabolism , Lipopolysaccharides/immunology , Lung/drug effects , Lung/immunology , Lung/pathology , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidation-Reduction/drug effects , Plant Proteins/isolation & purification , Plant Proteins/therapeutic use , Signal Transduction/immunology , Toll-Like Receptor 4/metabolism , Transcription Factor RelA/metabolism
11.
Food Funct ; 10(7): 4001-4009, 2019 Jul 17.
Article in English | MEDLINE | ID: mdl-31210194

ABSTRACT

Cinnamaldehyde is a flavor isolated from the bark of Cinnamomum cassia Presl and exerts anti-inflammation effects in various diseases. In our study, we investigated the protective effects and the potential mechanism of cinnamaldehyde on atherosclerosis (AS) by using a high fat diet (HFD)-fed ApoE-/- atherosclerotic mouse model. Here, we found that the serum LDL-C, TG and TC levels were elevated and the HDL-C level was decreased in HFD-fed ApoE-/- mice. Cinnamaldehyde treatment significantly decreased inflammatory cytokine (TNF-α, IL-6, NO and MCP-1) overproduction and the serum lipid level. Meanwhile, cinnamaldehyde increased the HDL-C level and down-regulated the activity of lipid peroxidation product MDA in serum. Moreover, cinnamaldehyde reduced the atherosclerotic plaque area in ApoE-/- mice. Furthermore, cinnamaldehyde reduced matrix metalloproteinase-2 (MMP-2) expression and attenuated the high phosphorylation level of IκBα and p65 NF-κB. Overall, our study indicates that cinnamaldehyde may achieve the anti-atherosclerotic effect via the IκB/NF-κB signaling pathway.


Subject(s)
Acrolein/analogs & derivatives , Apolipoproteins E/genetics , Atherosclerosis/drug therapy , Diet, High-Fat/adverse effects , I-kappa B Proteins/metabolism , NF-kappa B/metabolism , Signal Transduction/drug effects , Acrolein/pharmacology , Acrolein/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Aorta, Thoracic/pathology , Atherosclerosis/pathology , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Cinnamomum aromaticum/chemistry , Cytokines/metabolism , Disease Models, Animal , Male , Malondialdehyde/blood , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-KappaB Inhibitor alpha/metabolism , Nitric Oxide/blood , Phosphorylation , Plant Extracts/pharmacology , Plaque, Atherosclerotic/drug therapy , Transcription Factor RelA/metabolism
12.
Eur J Pharmacol ; 849: 160-169, 2019 Apr 15.
Article in English | MEDLINE | ID: mdl-30716318

ABSTRACT

Acute kidney injury is a common clinical condition associated with increased morbidity and mortality. It is essential to find effective drugs with low side effects in the treatment of acute kidney injury. Harmine is one of the major active components of Peganum harmala L. Harmine possesses various pharmacological activities, including anti-inflammatory activity. Nevertheless, the protective effect of harmine in acute kidney injury induced by lipopolysaccharide (LPS) in mice is unknown. Therefore, we investigated the protective effect of harmine in LPS-induced renal inflammation and the involved molecular mechanisms. The results showed that pretreatment with harmine (25 or 50 mg/kg) markedly alleviated kidney injury by reducing the release of kidney biomarkers and inflammatory mediators and the formation of malondialdehyde (MDA) and myeloperoxidase (MPO) while increasing superoxide dismutase (SOD) and glutathione (GSH) activities and improving renal histopathological changes. In addition, immunohistochemistry staining and western blot analysis indicated that harmine treatment suppressed the expression of toll-like receptor 4 (TLR4) and the phosphorylation of nuclear factor-kappa B (NF-κB) p65 and inhibitor of κBα (IκBα) while inhibiting the expression of NLRP3, caspase-1 and interleukin-1ß (IL-1ß). In brief, harmine protects against acute kidney injury induced by LPS in mice through reducing oxidative stress and inflammation responses. The involved underlying mechanisms of harmine in LPS-induced acute kidney injury might be related to inhibition of the TLR4-NF-κB pathway and NLRP3 inflammasome pathway. Based on the above conclusion, it is possible for harmine to be used to clinically treat acute kidney injury.


Subject(s)
Acute Kidney Injury/drug therapy , Harmine/pharmacology , Inflammasomes/metabolism , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Toll-Like Receptor 4/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Harmine/therapeutic use , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Male , Mice , Signal Transduction/drug effects
13.
Int Immunopharmacol ; 67: 1-12, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30530164

ABSTRACT

Fraxin, the effective component of the Chinese traditional medicine Cortex Fraxini, is reported to have anti-inflammatory effects. This study assessed the anti-inflammatory effect of fraxin on the lipopolysaccharide (LPS)-induced inflammatory response in A549 cells and the protective efficacy on LPS-induced acute lung injury (ALI) in mice. Fraxin reduced LPS-induced TNF-α, IL-6 and IL-1ß production in A549 cells and alleviated the LPS-induced wet/dry (W/D) weight ratio and the effects observed via histopathological examination of the lung in vivo. Furthermore, fraxin reduced the protein concentrations in the broncho-alveolar lavage (BAL) fluid and cytokine production in the sera. Fraxin also clearly attenuated the oxidation index, including the activity of myeloperoxidase (MPO), malondialdehyde (MDA), superoxide dismutase (SOD) and glutathione (GSH). Immunohistochemistry analysis showed that fraxin suppressed LPS-induced inflammatory damage. The expression of proteins involved in the NF-κB and NLRP3 inflammatory corpuscle signalling pathways was consistent between the lung tissues and cell samples. Overall, fraxin played a protective role in LPS-induced lung injury by inhibiting the NF-κB and NLRP3 signalling pathways.


Subject(s)
Acute Lung Injury/drug therapy , Anti-Inflammatory Agents/therapeutic use , Coumarins/therapeutic use , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Respiratory Mucosa/drug effects , A549 Cells , Acute Lung Injury/chemically induced , Animals , Cytokines/metabolism , Disease Models, Animal , Humans , Inflammation Mediators/metabolism , Lipopolysaccharides/immunology , Male , Medicine, Chinese Traditional , Mice , Mice, Inbred Strains , Oxidation-Reduction/drug effects , Peroxidase/metabolism , Respiratory Mucosa/pathology , Signal Transduction
14.
Acta Crystallogr Sect E Struct Rep Online ; 65(Pt 1): o75, 2008 Dec 10.
Article in English | MEDLINE | ID: mdl-21581714

ABSTRACT

In the title compound, C(26)H(20)N(6)O(4), the two aroylhydrazone side groups exist as diastereomeres, both in the keto form in the crystal structure. The aroylhydrazone units support the mol-ecular conformation through an intra-molecular N-H⋯O hydrogen bond. Two mol-ecules are connected into a centrosymmetric dimer by inter-molecular N-H⋯N hydrogen bonds. These dimers are connected into chains along the a axis by inter-molecular O-H⋯O hydrogen bonds. The combination of these hydrogen bonds results in layers in the bc plane. The layers are further linked by weak C-H⋯π contacts to form a three-dimensional network structure.

SELECTION OF CITATIONS
SEARCH DETAIL
...