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

Complementary Medicines
Publication year range
1.
Zhen Ci Yan Jiu ; 49(4): 331-340, 2024 Apr 25.
Article in English, Chinese | MEDLINE | ID: mdl-38649200

ABSTRACT

OBJECTIVES: To observe whether acupuncture up-regulates chemokine CXC ligand 1 (CXCL1) in the brain to play an analgesic role through CXCL1/chemokine CXC receptor 2 (CXCR2) signaling in adjuvant induced arthritis (AIA) rats, so as to reveal its neuro-immunological mechanism underlying improvement of AIA. METHODS: BALB/c mice with relatively stable thermal pain reaction were subjected to planta injection of complete Freund adjuvant (CFA) for establishing AIA model, followed by dividing the AIA mice into simple AF750 (fluorochrome) and AF750+CXCL1 groups (n=2 in each group). AF750 labeled CXCL1 recombinant protein was then injected into the mouse's tail vein to induce elevation of CXCL1 level in blood for simulating the effect of acupuncture stimulation which has been demonstrated by our past study. In vivo small animal imaging technology was used to observe the AF750 and AF750+CXCL1-labelled target regions. After thermal pain screening, the Wistar rats with stable pain reaction were subjected to AIA modeling by injecting CFA into the rat's right planta, then were randomized into model and manual acupuncture groups (n=12 in each group). Other 12 rats that received planta injection of saline were used as the control group. Manual acupuncture (uniform reinforcing and reducing manipulations) was applied to bilateral "Zusanli" (ST36) for 4×2 min, with an interval of 5 min between every 2 min, once daily for 7 days. The thermal pain threshold was assessed by detecting the paw withdrawal latency (PWL) using a thermal pain detector. The contents of CXCL1 in the primary somatosensory cortex (S1), medial prefrontal cortex, nucleus accumbens, amygdala, periaqueductal gray and rostroventromedial medulla regions were assayed by using ELISA, and the expression levels of CXCL1, CXCR2 and mu-opioid receptor (MOR) mRNA in the S1 region were detected using real time-quantitative polymerase chain reaction. The immune-fluorescence positive cellular rate of CXCL1 and CXCR2 in S1 region was observed after immunofluorescence stain. The immunofluorescence double-stain of CXCR2 and astrocyte marker glial fibrillary acidic protein (GFAP) or neuron marker NeuN or MOR was used to determine whether there is a co-expression between them. RESULTS: In AIA mice, results of in vivo experiments showed no obvious enrichment signal of AF750 or AF750+CXCL1 in any organ of the body, while in vitro experiments showed that there was a stronger fluorescence signal of CXCL1 recombinant protein in the brain. In rats, compared with the control group, the PWL from day 0 to day 7 was significantly decreased (P<0.01) and the expression of CXCR2 mRNA in the S1 region significantly increased in the model group (P<0.05), while in comparison with the model group, the PWL from day 2 to day 7, CXCL1 content, CXCR2 mRNA expression and CXCR2 content, and MOR mRNA expression in the S1 region were significantly increased in the manual acupuncture group (P<0.05, P<0.01). Immunofluorescence stain showed that CXCR2 co-stained with NeuN and MOR in the S1 region, indicating that CXCR2 exists in neurons and MOR-positive neurons but not in GFAP positive astrocytes. CONCLUSIONS: Acupuncture can increase the content of CXCL1 in S1 region, up-regulate CXCR2 on neurons in the S1 region and improve MOR expression in S1 region of AIA rats, which may contribute to its effect in alleviating inflammatory pain.


Subject(s)
Acupuncture Therapy , Arthritis, Experimental , Chemokine CXCL1 , Receptors, Interleukin-8B , Somatosensory Cortex , Animals , Humans , Male , Mice , Rats , Acupuncture Points , Arthritis, Experimental/therapy , Arthritis, Experimental/metabolism , Arthritis, Experimental/genetics , Chemokine CXCL1/metabolism , Chemokine CXCL1/genetics , Inflammation/therapy , Inflammation/metabolism , Inflammation/genetics , Mice, Inbred BALB C , Pain/metabolism , Pain/genetics , Pain Management , Rats, Wistar , Receptors, Interleukin-8B/metabolism , Receptors, Interleukin-8B/genetics , Signal Transduction , Somatosensory Cortex/metabolism
2.
Immunopharmacol Immunotoxicol ; 45(6): 692-700, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37358143

ABSTRACT

OBJECTIVE: Treatment with TNF-α inhibitors improve psoriasis with minimize/minor neutrophils infiltration and CXCL-1/8 expression in psoriatic lesions. However, the fine mechanism of TNF-α initiating psoriatic inflammation by tuning keratinocytes is unclear. Our previous research identified the deficiency of intracellular galectin-3 was sufficient to promote psoriasis inflammation characterized by neutrophil accumulation. This study aims to investigate whether TNF-α participated in psoriasis development through dysregulating galectin-3 expression. METHODS: mRNA levels were assessed through quantitative real-time PCR. Flow cytometry was used to detect cell cycle/apoptosis. Western blot was used to evaluate the activation of the NF-κB signaling pathway. HE staining and immunochemistry were used to detect epidermal thickness and MPO expression, respectively. Specific small interfering RNA (siRNA) was used to knock down hsa-miR-27a-3p while plasmids transfection was used to overexpress galectin-3. Further, the multiMiR R package was utilized to predict microRNA-target interaction. RESULTS AND DISCUSSION: We found that TNF-α stimulation altered cell proliferation and differentiation and promoted the production of psoriasis-related inflammatory mediators along with the inhibition of galectin-3 expression in keratinocytes. Supplement of galectin-3 could counteract the rise of CXCL-1/8 but not the other phenotypes of keratinocytes induced by TNF-α. Mechanistically, inhibition of the NF-κB signaling pathway could counteract the decrease of galectin-3 and the increase of hsa-miR-27a-3p expression whereas silence of hsa-miR-27a-3p could counteract the decrease of galectin-3 expression induced by TNF-α treatment in keratinocytes. Intradermal injection of murine anti-CXCL-2 antibody greatly alleviated imiquimod-induced psoriasis-like dermatitis. CONCLUSION: TNF-α initiates psoriatic inflammation by increasing CXCL-1/8 in keratinocytes mediated by the axis of NF-κB-hsa-miR-27a-3p-galectin-3 pathway.


Subject(s)
Galectin 3 , Keratinocytes , MicroRNAs , Psoriasis , Tumor Necrosis Factor-alpha , Tumor Necrosis Factor-alpha/pharmacology , Keratinocytes/metabolism , HaCaT Cells , Humans , MicroRNAs/genetics , Chemokine CXCL1/metabolism , Interleukin-8/metabolism , Galectin 3/genetics , Psoriasis/genetics , Psoriasis/pathology , NF-kappa B/metabolism , Signal Transduction , Female , Animals , Mice , Mice, Inbred C57BL
3.
Phytomedicine ; 90: 153628, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34247114

ABSTRACT

BACKGROUND: Metastasis is the most common lethal cause of breast cancer-related death. Recent studies have implied that autophagy is closely implicated in cancer metastasis. Therefore, it is of great significance to explore autophagy-related molecular targets involved in breast cancer metastasis and to develop therapeutic drugs. PURPOSE: This study was designed to investigate the anti-metastatic effects and autophagy regulatory mechanisms of Aiduqing (ADQ) formula on breast cancer. STUDY DESIGN/METHODS: Multiple cellular and molecular experiments were conducted to investigate the inhibitory effects of ADQ formula on autophagy and metastasis of breast cancer cells in vitro. Meanwhile, autophagic activator/inhibitor as well as CXCL1 overexpression or interference plasmids were used to investigate the underlying mechanisms of ADQ formula in modulating autophagy-mediated metastasis. Furthermore, the zebrafish xenotransplantation model and mouse xenografts were applied to validate the inhibitory effect of ADQ formula on autophagy-mediated metastasis in breast cancer in vivo. RESULTS: ADQ formula significantly inhibited the proliferation, migration, invasion and autophagy but induced apoptosis of high-metastatic breast cancer cells in vitro. Similar results were also observed in starvation-induced breast cancer cells which exhibited elevated metastatic ability and autophagy activity. Mechanism investigations further approved that either CXCL1 overexpression or autophagic activator rapamycin can significantly abrogated the anti-metastatic effects of ADQ formula, suggesting that CXCL1-mediated autophagy may be the crucial pathway of ADQ formula in suppressing breast cancer metastasis. More importantly, ADQ formula suppressed breast cancer growth, autophagy, and metastasis in both the zebrafish xenotransplantation model and the mouse xenografts. CONCLUSION: Our study not only revealed the novel function of CXCL1 in mediating autophagy-mediated metastasis but also suggested ADQ formula as a candidate drug for the treatment of metastatic breast cancer.


Subject(s)
Autophagy/drug effects , Breast Neoplasms , Chemokine CXCL1/metabolism , Drugs, Chinese Herbal/pharmacology , Neoplasm Metastasis/prevention & control , Animals , Apoptosis , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation , Female , Humans , Mice , Xenograft Model Antitumor Assays , Zebrafish
4.
Front Endocrinol (Lausanne) ; 12: 601160, 2021.
Article in English | MEDLINE | ID: mdl-33815271

ABSTRACT

Background: Non-alcoholic fatty liver disease (NAFLD) is a complex multifactorial disorder that is characterised by dysfunctional lipid metabolism and cholesterol homeostasis, and a related chronic inflammatory response. NAFLD has become the most common cause of chronic liver disease in many countries, and its prevalence continues to rise in parallel with increasing rates of obesity. Here, we evaluated the putative NAFLD-attenuating effects of a multicomponent medicine consisting of 24 natural ingredients: Hepar compositum (HC-24). Methods: Ldlr-/-.Leiden mice were fed a high-fat diet (HFD) with a macronutrient composition and cholesterol content comparable to human diets for 24 weeks to induce obesity-associated metabolic dysfunction, including hepatic steatosis and inflammation. HC-24 or vehicle control was administered intraperitoneally 3 times/week (1.5 ml/kg) for the last 18 weeks of the study. Histological analyses of liver and adipose tissue were combined with extensive hepatic transcriptomics analysis. Transcriptomics results were further substantiated with ELISA, immunohistochemical and liver lipid analyses. Results: HFD feeding induced obesity and metabolic dysfunction including adipose tissue inflammation and increased gut permeability. In the liver, HFD-feeding resulted in a disturbance of cholesterol homeostasis and an associated inflammatory response. HC-24 did not affect body weight, metabolic risk factors, adipose tissue inflammation or gut permeability. While HC-24 did not alter total liver steatosis, there was a pronounced reduction in lobular inflammation in HC-24-treated animals, which was associated with modulation of genes and proteins involved in inflammation (e.g., neutrophil chemokine Cxcl1) and cholesterol homeostasis (i.e., predicted effect on 'cholesterol' as an upstream regulator, based on gene expression changes associated with cholesterol handling). These effects were confirmed by CXCL1 ELISA, immunohistochemical staining of neutrophils and biochemical analysis of hepatic free cholesterol content. Intrahepatic free cholesterol levels were found to correlate significantly with the number of inflammatory aggregates in the liver, thereby providing a potential rationale for the observed anti-inflammatory effects of HC-24. Conclusions: Free cholesterol accumulates in the liver of Ldlr-/-.Leiden mice under physiologically translational dietary conditions, and this is associated with the development of hepatic inflammation. The multicomponent medicine HC-24 reduces accumulation of free cholesterol and has molecular and cellular anti-inflammatory effects in the liver.


Subject(s)
Cholesterol/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Plant Extracts/administration & dosage , Animals , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Diet, High-Fat/adverse effects , Humans , Lipid Metabolism/drug effects , Liver/drug effects , Liver/immunology , Liver/metabolism , Male , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/metabolism , Receptors, LDL/genetics , Receptors, LDL/immunology
5.
FASEB J ; 35(4): e21354, 2021 04.
Article in English | MEDLINE | ID: mdl-33749892

ABSTRACT

ω3 fatty acids show potent bioactivities via conversion into lipid mediators; therefore, metabolism of dietary lipids is a critical determinant in the properties of ω3 fatty acids in the control of allergic inflammatory diseases. However, metabolic progression of ω3 fatty acids in the skin and their roles in the regulation of skin inflammation remains to be clarified. In this study, we found that 12-hydroxyeicosapentaenoic acid (12-HEPE), which is a 12-lipoxygenase metabolite of eicosapentaenoic acid, was the prominent metabolite accumulated in the skin of mice fed ω3 fatty acid-rich linseed oil. Consistently, the gene expression levels of Alox12 and Alox12b, which encode proteins involved in the generation of 12-HEPE, were much higher in the skin than in the other tissues (eg, gut). We also found that the topical application of 12-HEPE inhibited the inflammation associated with contact hypersensitivity by inhibiting neutrophil infiltration into the skin. In human keratinocytes in vitro, 12-HEPE inhibited the expression of two genes encoding neutrophil chemoattractants, CXCL1 and CXCL2, via retinoid X receptor α. Together, the present results demonstrate that the metabolic progression of dietary ω3 fatty acids differs in different organs, and identify 12-HEPE as the dominant ω3 fatty acid metabolite in the skin.


Subject(s)
Chemokine CXCL1/metabolism , Dermatitis, Contact/prevention & control , Eicosapentaenoic Acid/analogs & derivatives , Keratinocytes/drug effects , Animals , Antibodies, Monoclonal/drug effects , Antibodies, Monoclonal/metabolism , Bone Marrow Cells , Chemokine CXCL1/genetics , Diet , Dinitrofluorobenzene , Down-Regulation , Eicosapentaenoic Acid/pharmacology , Female , Gene Expression Regulation/drug effects , HaCaT Cells , Humans , Linseed Oil/administration & dosage , Linseed Oil/metabolism , Mice
6.
Res Q Exerc Sport ; 92(1): 82-90, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32109201

ABSTRACT

Purpose: This study aimed to evaluate whether supplementation with L-arginine alone or in combination with physical exercise training can modulate rats' lipid and inflammatory profiles after a single intense exercise session. Methods: Male Wistar rats were divided into four different groups: control (C), trained (T), supplemented with L-arginine (C + A) and trained and supplemented (T + A). Animals from supplemented groups (C + A and T + A groups) received 300 mg/kg animal body weight L-arginine diluted in 30 mL of drinking water for 8 weeks. Exercise training protocol (moderate intensity-70% achieved in the maximum effort test) was held 5 days/week for 8 weeks. Results: Exercise training induced a decrease in the amount of plasma, cholesterol and triglyceride totals, and skeletal muscle VEGF and CINC. Supplementation alone showed a benefit by reducing LDL levels. Conclusion: Training combined with supplementation showed a pronounced reduction in skeletal muscle VEGF and CINC amount. L-arginine supplementation, especially when associated with the regular aerobic physical exercise at moderate intensity was able to improve not only plasma lipid profile but also the inflammatory response of skeletal muscle immediately after an exhaustive physical exercise session.


Subject(s)
Arginine/administration & dosage , Dietary Supplements , Lipids/blood , Muscle, Skeletal/metabolism , Myositis/metabolism , Physical Conditioning, Animal/physiology , Animals , Chemokine CXCL1/metabolism , Cytokines/metabolism , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/injuries , Rats, Wistar , Vascular Endothelial Growth Factor A/metabolism
7.
Phytomedicine ; 78: 153331, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32911383

ABSTRACT

BACKGROUND: Breast cancer is the most common malignancy in women and metastasis is the leading cause of breast cancer-related deaths. Our previous studies have shown that XIAOPI formula, a newly approved drug by the State Food and Drug Administration of China (SFDA), can dramatically inhibit breast cancer metastasis by modulating the tumor-associated macrophages/C-X-C motif chemokine ligand 1 (TAMs/CXCL1) pathway. However, the bioactive compound accounting for the anti-metastatic effect of XIAOPI formula remains unclear. PURPOSE: This study was designed to separate the anti-metastatic bioactive compound from XIAOPI formula and to elucidate its action mechanisms. STUDY DESIGN/METHODS: TAMs/CXCL1 promoter activity-guided fractionation and multiple chemical structure identification approaches were conducted to screen the bioactive compound from XIAOPI formula. Breast cancer cells and TAMs were co-cultured in vitro or co-injected in vivo to simulate their coexistence. Multiple molecular biology experiments, zebrafish breast cancer xenotransplantation model and mouse breast cancer xenografts were applied to validate the anti-metastatic activity of the screened compound. RESULTS: Bioactivity-guided fractionation identified baohuoside I (BHS) as the key bioactive compound of XIAOPI formula in inhibiting TAMs/CXCL1 promoter activity. Functional studies revealed that BHS could significantly inhibit the migration and invasion as well as the expression of metastasis-related proteins in both human and mouse breast cancer cells, along with decreasing the proportion of breast cancer stem cells (CSCs). Furthermore, BHS could suppress the M2 phenotype polarization of TAMs and therefore attenuate their CXCL1 expression and secretion. Notably, mechanistic investigations validated TAMs/CXCL1 as the crucial target of BHS in suppressing breast cancer metastasis as exogenous addition of CXCL1 significantly abrogated the anti-metastatic effect of BHS on breast cancer cells. Moreover, BHS was highly safe in vivo as it exhibited no observable embryotoxicity or teratogenic effect on zebrafish embryos. More importantly, BHS remarkably suppressed breast cancer metastasis and TAMs/CXCL1 activity in both zebrafish breast cancer xenotransplantation model and mouse breast cancer xenografts. CONCLUSION: This study not only provides novel insights into TAMs/CXCL1 as a reliable screening target for anti-metastatic drug discovery, but also suggests BHS as a promising candidate drug for metastatic breast cancer treatment.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Chemokine CXCL1/metabolism , Flavonoids/pharmacology , Macrophages/drug effects , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Chemokine CXCL1/antagonists & inhibitors , Chemokine CXCL1/genetics , Coculture Techniques , Down-Regulation/drug effects , Drug Screening Assays, Antitumor , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Embryo, Nonmammalian , Female , Humans , Macrophages/metabolism , Macrophages/pathology , Mice, Inbred BALB C , Neoplasm Metastasis , Neoplastic Stem Cells/drug effects , Xenograft Model Antitumor Assays , Zebrafish/embryology
8.
Cell Commun Signal ; 18(1): 48, 2020 03 26.
Article in English | MEDLINE | ID: mdl-32213179

ABSTRACT

BACKGROUND: Recent findings suggested that premetastatic niche (PMN) is a prerequisite in mediating cancer metastasis. Previously we demonstrated that XIAOPI formula could inhibit breast cancer lung metastasis via inhibiting tumor associated macrophages (TAMs)-secreted CXCL1. Herein, we aimed to explore the effects of XIAOPI formula on preventing breast cancer PMN formation and its underlying molecular mechanisms. METHODS: CXCL1 expression of TAMs was detected by qPCR and Western blotting assay. The influences of XIAOPI formula on the proliferation of TAMs and 4 T1 in the co-culture system were tested by CCK8 or EdU staining. Transwell experiment was applied to determine the effects of XIAOPI formula on the invasion ability of HSPCs and 4 T1. Breast cancer xenografts were built by inoculating 4 T1 cells into the mammary pads of Balb/c mice and lung metastasis was monitored by luciferase imaging. Immune fluorescence assay was used to test the epithelial-mesenchymal transition process and PMN formation in the lung tissues. The effects of XIAOPI formula on TAMs phenotype, hematopoietic stem/progenitor cells (HSPCs) and myeloid-derived suppressor cells (MDSCs) were determined by flow cytometry. RESULTS: It was found that XIAOPI formula could inhibit the proliferation and polarization of M2 phenotype macrophages, and reduce CXCL1 expression in a dose-dependent manner. However, M1 phenotype macrophages were not significantly affected by XIAOPI formula. TAMs/CXCL1 signaling was subsequently found to stimulate the recruitment of c-Kit+/Sca-1+ HSPCs and their differentiation into CD11b+/Gr-1+ MDSCs, which were symbolic events accounting for PMN formation. Moreover, XIAOPI formula was effective in inhibiting HSPCs activation and suppressing the proliferation and metastasis of breast cancer cells 4 T1 induced by HSPCs and TAMs co-culture system, implying that XIAOPI was effective in preventing PMN formation in vitro. Breast cancer xenograft experiments further demonstrated that XIAOPI formula could inhibit breast cancer PMN formation and subsequent lung metastasis in vivo. The populations of HSPCs in the bone marrow and MDSCs in the lung tissues were all remarkably declined by XIAOPI formula treatment. However, the inhibitory effects of XIAOPI formula could be relieved by CXCL1 overexpression in the TAMs. CONCLUSIONS: Taken together, our study provided preclinical evidence supporting the application of XIAOPI formula in preventing breast cancer PMN formation, and highlighted TAMs/CXCL1 as a potential therapeutic strategy for PMN targeting therapy. Video Abstract.


Subject(s)
Breast Neoplasms , Chemokine CXCL1/metabolism , Lung Neoplasms , Plant Extracts , Tumor-Associated Macrophages/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Proliferation , Female , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Plant Extracts/administration & dosage , Plant Extracts/pharmacology , RAW 264.7 Cells , Tumor Microenvironment , Tumor-Associated Macrophages/cytology
9.
Thorax ; 75(3): 253-261, 2020 03.
Article in English | MEDLINE | ID: mdl-31915307

ABSTRACT

INTRODUCTION: Zinc is well known for its anti-inflammatory effects, including regulation of migration and activity of polymorphonuclear neutrophils (PMN). Zinc deficiency is associated with inflammatory diseases such as acute lung injury (ALI). As deregulated neutrophil recruitment and their hyper-activation are hallmarks of ALI, benefits of zinc supplementation on the development of lipopolysaccharides (LPS)-induced ALI were tested. METHODS: 64 C57Bl/6 mice, split into eight groups, were injected with 30 µg zinc 24 hours before exposure to aerosolised LPS for 4 hours. Zinc homoeostasis was characterised measuring serum and lung zinc concentrations as well as metallothionein-1 expression. Recruitment of neutrophils to alveolar, interstitial and intravascular space was assessed using flow cytometry. To determine the extent of lung damage, permeability and histological changes and the influx of protein into the bronchoalveolar lavage fluid were measured. Inflammatory status and PMN activity were evaluated via tumour necrosis factor α levels and formation of neutrophil extracellular traps. The effects of zinc supplementation prior to LPS stimulation on activation of primary human granulocytes and integrity of human lung cell monolayers were assessed as well. RESULTS: Injecting zinc 24 hours prior to LPS-induced ALI indeed significantly decreased the recruitment of neutrophils to the lungs and prevented their hyperactivity and thus lung damage was decreased. Results from in vitro investigations using human cells suggest the transferability of the finding to human disease, which remains to be tested in more detail. CONCLUSION: Zinc supplementation attenuated LPS-induced lung injury in a murine ALI model. Thus, the usage of zinc-based strategies should be considered to prevent detrimental consequences of respiratory infection and lung damage in risk groups.


Subject(s)
Acute Lung Injury/metabolism , Acute Lung Injury/prevention & control , Neutrophil Infiltration/drug effects , Neutrophils/physiology , Zinc/pharmacology , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Cation Transport Proteins/genetics , Cell Line , Cell Survival/drug effects , Chemokine CXCL1/metabolism , Disease Models, Animal , Gene Expression/drug effects , Granulocyte Colony-Stimulating Factor/genetics , Homeostasis , Humans , L-Selectin/metabolism , Lipopolysaccharides , Male , Metallothionein/genetics , Metallothionein/metabolism , Mice , Mice, Inbred C57BL , RNA, Messenger , Receptors, Complement 3b/metabolism , STAT3 Transcription Factor/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Zinc/metabolism , Zinc/therapeutic use
10.
Oxid Med Cell Longev ; 2020: 7574606, 2020.
Article in English | MEDLINE | ID: mdl-33628359

ABSTRACT

BACKGROUND: Pulicaria crispa (P. crispa) is a plant from the Compositae family that exhibits antioxidant, anti-inflammatory, antibacterial, and cytotoxic activities. OBJECTIVE: The current study aimed at investigating the immunomodulatory effects of P. crispa extract in lipopolysaccharide- (LPS-) stimulated human monocytic THP-1 cells. METHODS: To induce macrophage differentiation, THP-1 cell lines were treated with phorbol-12-myristate 13-acetate, followed by exposure to LPS with or without 50 or 100 µg/ml of P. crispa extract. The following tests were employed to test the immunomodulatory effects of the extract: MTT assay, ELISA, Western blotting analysis, cell migration and phagocytosis assays, and Annexin V staining method. RESULTS: Exposure to 100 µg/ml P. crispa extract significantly reduced THP-1 cell proliferation, migration, and phagocytosis (in LPS-stimulated cells, but not in unstimulated cells). Moreover, the extract alone significantly reduced the rate of THP-1 cell apoptosis, while it increased the rate of late apoptosis. Molecular investigations showed that treatment with P. crispa extract significantly upregulated the expression of ERK1, p-MAPK, P-P38, and Bcl2, while it significantly reduced the expression of ERK5, Bax, NF-κB, P-NF-κB, CCL1, CCL2, CCL5, CCL22, CXCL1, and CXCL10. CONCLUSION: Pulicaria crispa extract exhibited anti-inflammatory, antiproliferative, antimigratory, and antiphagocytic effects in LPS-stimulated THP-1 cells. Future studies should investigate these mechanisms in animal models with chronic inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Immunomodulation/drug effects , Monocytes/drug effects , Plant Extracts/pharmacology , Pulicaria/chemistry , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Chemokine CXCL1/metabolism , Chemokines, CC/metabolism , Down-Regulation , Humans , Lipopolysaccharides/pharmacology , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/metabolism , Phagocytosis/drug effects , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/metabolism , Pulicaria/metabolism , THP-1 Cells , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/pharmacology , Up-Regulation , bcl-2-Associated X Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Inflammopharmacology ; 28(1): 201-213, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31388880

ABSTRACT

Some B vitamins exhibit activities in models of nociceptive pain, inflammatory pain, and neuropathic pain induced by nerve lesions and also in certain painful conditions in humans. In the present study, we investigated the effects of thiamine, riboflavin, and nicotinamide in a neuropathic pain model induced by the chemotherapeutic paclitaxel in mice. Four intraperitoneal (i.p.) administrations of paclitaxel (2 mg/kg day, cumulative dose 8 mg/kg) induced a long-lasting mechanical allodynia. Per os (p.o.) administration of two doses of thiamine (150, 300 and 600 mg/kg), nicotinamide (250, 500 and 1000 mg/kg) or riboflavin (125, 250 and 500 mg/kg), on the seventh day after the first administration of paclitaxel, the mechanical allodynia was attenuated. The antinociceptive activity of all B vitamins was attenuated by glibenclamide (20 and 10 mg/kg, p.o.). Naltrexone (5 and 10 mg/kg, i.p.) attenuated the antinociceptive activity of thiamine. Thiamine, riboflavin, and nicotinamide also reduced the concentrations of tumor necrosis factor-α (TNF-α) and CXCL-1 in dorsal root ganglia (DRG) and thalamus. In conclusion, thiamine, riboflavin, and nicotinamide exhibit antinociceptive activity in the neuropathic pain model induced by paclitaxel. Inhibition of TNF-α and CXCL-1 production in DRG and thalamus, as well as activation of ATP-sensitive potassium channels, underly their antinociceptive activity.


Subject(s)
Chemokine CXCL1/metabolism , Ganglia, Spinal/drug effects , Hyperalgesia/drug therapy , KATP Channels/metabolism , Thalamus/drug effects , Tumor Necrosis Factor-alpha/metabolism , Vitamin B Complex/pharmacology , Animals , Ganglia, Spinal/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Male , Mice , Neuralgia/drug therapy , Neuralgia/metabolism , Niacinamide/pharmacology , Paclitaxel/pharmacology , Riboflavin/pharmacology , Thalamus/metabolism , Thiamine/pharmacology
12.
Biomed Pharmacother ; 120: 109519, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31629951

ABSTRACT

XIAOPI formula is a national approved drug prescribed to patients with high breast cancer risk. Previously we demonstrated that XIAOPI formula could inhibit breast cancer metastasis via suppressing CXCL1 expression, and postulated that "autophagy in cancer" might be one of its most core anti-cancer mechanisms. However, whether XIAOPI formula could be simultaneously applied with chemodrugs and their synergistic mechanisms are still remained unknown. In the present study, XIAOPI formula at non-cytotoxic doses could synergistically enhance the chemosensitivity of breast cancer cells MDA-MB-231 and MCF-7. We found that rapamycin-induced autophagy could reduce the chemosensitivity of breast cancer cells to XIAOPI formula, and the autophagy suppression and chemosensitizing activity of this formula was CXCL1-dependent. The evidence came from that XIAOPI formula was associated with a lower expression of CXCL1 combined with either rapamycin or taxol alone. Besides, the inhibitory effect of XIAOPI formula on the LC3-II and ABCG2 signals was weakened following CXCL1 over-expression, whereas P62 upregulation induced by XIAOPI formula was re-declined. A high throughput - qPCR (HT-qPCR) assay identified HMGB1 as the main autophagic target of XIAOPI formula in chemosensitizing breast cancer. and furhter validation suggested XIAOPI formula exerted chemosensitivity mainly via CXCL1/HMGB1 autophagic axis. Finally, we generated both mice and zebrafish xenotransplantation models bearing MDA-MB-231 breast cancer cells, and found that XIAOPI formula safely enhanced in vivo taxol chemosensitivity on breast cancer. Taken together, XIAOPI formula is a potential adjuvant drug via inhibiting CXCL1/HMGB1-mediated autophagy for breast cancer treatment with good safety.


Subject(s)
Breast Neoplasms/drug therapy , Chemokine CXCL1/metabolism , Drug Resistance, Neoplasm/drug effects , Drugs, Chinese Herbal/pharmacology , HMGB1 Protein/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Autophagy , Cell Line , Cell Survival , Chemokine CXCL1/genetics , Drug Synergism , Epirubicin/metabolism , Female , Gene Expression Regulation/drug effects , HMGB1 Protein/genetics , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Nude , Neoplasms, Experimental/drug therapy , Oviposition/drug effects , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Zebrafish
13.
Physiol Rep ; 7(18): e14235, 2019 09.
Article in English | MEDLINE | ID: mdl-31552707

ABSTRACT

l-homoarginine is an endogenous, non-proteinogenic amino acid that has emerged as a new player in health and disease. Specifically, low l-homoarginine levels are associated with cardiovascular diseases, stroke, and reduced kidney function. However, the role of l-homoarginine in the pathogenesis of diabetic nephropathy (DN) is not known. Experiments were conducted in 6-week-old Ins2Akita mice supplemented with l-homoarginine via drinking water or mini osmotic pump for 12 weeks. Both plasma and kidney l-homoarginine levels were significantly reduced in diabetic mice compared to nondiabetic controls. Untreated Ins2Akita mice showed significant increases in urinary albumin excretion, histological changes, glomerular macrophage recruitment, the inflammatory cytokine KC-GRO/CXCL1, and urinary thiobarbituric acid reactive substances (TBARS) excretion as an indicator of oxidative stress, along with a significant reduction in kidney nitrate + nitrite levels compared to control mice at 18 weeks of age. In contrast, l-homoarginine supplementation for 12 weeks in Ins2Akita mice, via either drinking water or mini osmotic pump, significantly reduced albuminuria, renal histological changes, glomerular macrophage recruitment, KC-GRO/CXCL1 levels, urinary TBARS excretion, and largely restored kidney nitrate + nitrite levels. These data demonstrate that l-homoarginine supplementation attenuates specific features of DN in mice and could be a potential new therapeutic tool for treating diabetic patients.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Diabetic Nephropathies/prevention & control , Dietary Supplements , Homoarginine/therapeutic use , Albuminuria/drug therapy , Animals , Chemokine CXCL1/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Drug Evaluation, Preclinical/methods , Homoarginine/metabolism , Kidney/metabolism , Kidney/pathology , Kidney Glomerulus/pathology , Macrophages/pathology , Mice, Inbred DBA , Nitrates/metabolism , Nitrites/metabolism , Oxidative Stress/drug effects
14.
Sci Rep ; 9(1): 6071, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30988402

ABSTRACT

We investigated the anti-inflammatory and antibacterial activities of Hc-cath, a cathelicidin peptide derived from the venom of the sea snake, Hydrophis cyanocyntus, using in vivo models of inflammation and infection. Hc-cath function was evaluated in in vitro, in vivo in the wax moth, Galleria mellonella, and in mouse models of intraperitoneal and respiratory Pseudomonas aeruginosa infection. Hc-Cath downregulated LPS-induced pro-inflammatory responses in macrophages and significantly improved the survival of P. aeruginosa infected G. mellonella over a 5-day period. We also demonstrated, for the first time, that Hc-cath can modulate inflammation in a mouse model of LPS-induced lung inflammation by significantly reducing the release of the pro-inflammatory cytokine and neutrophil chemoattractant, KC, resulting in reduced cellular infiltration into the lungs. Moreover, Hc-cath treatment significantly reduced the bacterial load and inflammation in mouse models of P. aeruginosa intraperitoneal and respiratory infection. The effect of Hc-cath in our studies highlights the potential to develop this peptide as a candidate for therapeutic development.


Subject(s)
Anti-Infective Agents/administration & dosage , Antimicrobial Cationic Peptides/administration & dosage , Biological Products/administration & dosage , Hydrophiidae , Pneumonia/drug therapy , Pseudomonas Infections/drug therapy , Animals , Anti-Infective Agents/chemical synthesis , Antimicrobial Cationic Peptides/chemical synthesis , Bacterial Load/drug effects , Bacterial Load/immunology , Biological Products/chemical synthesis , Chemokine CXCL1/immunology , Chemokine CXCL1/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Humans , Lipopolysaccharides/immunology , Lung/drug effects , Lung/immunology , Lung/microbiology , Mice , Moths/immunology , Moths/microbiology , Pneumonia/immunology , Pneumonia/microbiology , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/isolation & purification , THP-1 Cells , Cathelicidins
15.
Pflugers Arch ; 471(2): 301-311, 2019 02.
Article in English | MEDLINE | ID: mdl-30349936

ABSTRACT

The classic model of fever induction is based on the administration of lipopolysaccharide (LPS) from Gram-negative bacteria in experimental animals. LPS-induced fever results in the synthesis/release of many mediators that assemble an LPS-fever cascade. We have previously demonstrated that cytokine-induced neutrophil chemoattractant (CINC)-1, a Glu-Leu-Arg (ELR) + chemokine, centrally administered to rats, induces fever and increases prostaglandin E2 in the cerebrospinal fluid. We now attempt to investigate the involvement of CINC-1 and its functional receptor CXCR2 on the fever induced by exogenous and endogenous pyrogens in rats. We also investigated the effect of reparixin, an allosteric inhibitor of CXCR1/CXCR2 receptors, on fever induced by either systemic administration of LPS or intracerebroventricular injection of CINC-1, as well as TNF-α, IL-1ß, IL-6, or ET-1, known mediators of febrile response. Our results show increased CINC-1 mRNA expression in the liver, hypothalamus, CSF, and plasma following LPS injection. Moreover, reparixin administered right before CINC-1 or LPS abolished the fever induced by CINC-1 and significantly reduced the response induced by LPS. In spite of these results, reparixin does not modify the fever induced by IL-1ß, TNF-α, and IL-6, but significantly reduces ET-1-induced fever. Therefore, it is plausible to suggest that CINC-1 might contribute to LPS-induced fever in rats by activating CXCR2 receptor on the CNS. Moreover, it can be hypothesized that CINC-1 is placed upstream TNF-α, IL-1ß, and IL-6 among the prostaglandin-dependent fever-mediator cascade and amidst the prostaglandin-independent synthesis pathway of fever.


Subject(s)
Chemokine CXCL1/metabolism , Fever/chemically induced , Fever/metabolism , Lipopolysaccharides/pharmacology , Receptors, Interleukin-8B/metabolism , Animals , Cerebrospinal Fluid/drug effects , Cerebrospinal Fluid/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Liver/drug effects , Liver/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Prostaglandins/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/metabolism
16.
J Pediatr Gastroenterol Nutr ; 68(2): 190-198, 2019 02.
Article in English | MEDLINE | ID: mdl-30247422

ABSTRACT

BACKGROUND: Enteroaggregative Escherichia coli (EAEC) is an important pathogen causing enteric infections worldwide. This pathotype is linked to malnutrition in children from developing countries. Alanyl-glutamine (Ala-Gln) is an immune modulator nutrient that acts during intestinal damage and/or inflammation. This study investigated the effect of EAEC infection and Ala-Gln on cell viability, cell death, and inflammation of intestinal epithelium cells (IEC-6). METHODS: Cells were infected with an EAEC prototype 042 strain, an EAEC wild-type strain isolated from a Brazilian malnourished child, and a commensal E coli HS. Gene transcription and protein levels of caspases-3, -8, and -9 and cytokine-induced neutrophil chemoattractant 1 (CINC-1/CXCL1) were evaluated using RT-qPCR, western blot analysis, and ELISA. RESULTS: Infections with both EAEC strains decreased cell viability and induced apoptosis and necrosis after 24 hours. Ala-Gln supplementation increased cell proliferation and reduced cell death in infected cells. Likewise, EAEC strain 042 significantly increased the transcript levels of caspases-3, -8, and -9 when compared to the control group, and Ala-Gln treatment reversed this effect. Furthermore, EAEC induced CXCL1 protein levels, which were also reduced by Ala-Gln supplementation. CONCLUSION: These findings suggest that EAEC infection promotes apoptosis, necrosis, and intestinal inflammation with involvement of caspases. Supplementation of Ala-Gln inhibits cell death, increases cell proliferation, attenuates mediators associated with cell death, and inflammatory pathways in infected cells.


Subject(s)
Cell Survival/drug effects , Dipeptides/pharmacology , Escherichia coli Infections/therapy , Escherichia coli/metabolism , Protective Agents/pharmacology , Chemokine CXCL1/metabolism , Child , Dietary Supplements , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/microbiology
17.
Nat Commun ; 9(1): 4613, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30397205

ABSTRACT

Many pro-inflammatory pathways leading to arthritis have global effects on the immune system rather than only acting locally in joints. The reason behind the regional and patchy distribution of arthritis represents a longstanding paradox. Here we show that biomechanical loading acts as a decisive factor in the transition from systemic autoimmunity to joint inflammation. Distribution of inflammation and erosive disease is confined to mechano-sensitive regions with a unique microanatomy. Curiously, this pathway relies on stromal cells but not adaptive immunity. Mechano-stimulation of mesenchymal cells induces CXCL1 and CCL2 for the recruitment of classical monocytes, which can differentiate into bone-resorbing osteoclasts. Genetic ablation of CCL2 or pharmacologic targeting of its receptor CCR2 abates mechanically-induced exacerbation of arthritis, indicating that stress-induced chemokine release by mesenchymal cells and chemo-attraction of monocytes determines preferential homing of arthritis to certain hot spots. Thus, mechanical strain controls the site-specific localisation of inflammation and tissue damage in arthritis.


Subject(s)
Arthritis/metabolism , Arthritis/pathology , Inflammation/metabolism , Adult , Animals , Arthritis/diagnostic imaging , Arthritis/genetics , Autoantibodies/metabolism , Autoimmunity , Bone Resorption/metabolism , Chemokine CXCL1/metabolism , Chemokine CXCL2/metabolism , Chemokines/metabolism , Disease Models, Animal , Female , Gene Expression , Humans , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Middle Aged , Monocytes , Osteoclasts/metabolism , Receptors, CCR2/drug effects , Stromal Cells , Tarsal Bones/diagnostic imaging , Tarsal Bones/pathology , Tendinopathy/pathology , Tendons/metabolism , X-Ray Microtomography
18.
Int J Mol Med ; 42(6): 3220-3230, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30272314

ABSTRACT

Systemic lupus erythematosus (SLE) is associated with an increased risk of vascular complications. Lupus nephritis is a major manifestation of SLE in the clinic. Lupus nephritis is elevated by T helper type 17 (Th17) cells, the major pro­inflammatory T­cell subset, leading to autoimmunity modulation. Therapeutic treatments targeting leukocyte recruitment may be useful in attenuating vascular complications linked to SLE progression. 3,7,3',4'­Tetrahydroxyflavone (fisetin) is a flavonol and a member of the flavonoid polyphenols. It is present in various fruits and vegetables, including persimmons, apples, kiwis, grapes, onions, strawberries and cucumbers. In the present study, the effects of fisetin against SLE induced by pristane (PRI) were evaluated in mice. Fisetin was indicated to reduce PRI­induced anti­double stranded DNA, anti­ small nuclear ribonucleoprotein and the ratio of albumin to creatinine in urine. In addition, the chemokine (C­X­C motif) ligand (CXCL) signaling pathway was activated for PRI treatment, which was reversed by fisetin administration by reducing CXCL­1 and 2, chemokine (C­C motif) ligand 3, as well as CXC receptor 2 expression. In addition, the induction of inflammatory cytokines, including interleukin (IL)­6, tumor necrosis factor­α, IL­1ß, as well as the chemokine interferon­Î³, by PRI were downregulated by fisetin treatment in mice. Furthermore, Th17 cells and their associated cytokines were highly induced by PRI treatment, which was inhibited by fisetin administration. The present results indicated that fisetin may be an effective management for SLE by targeting the CXCL signaling pathway and regulating Th17 differentiation during lupus nephritis development.


Subject(s)
Flavonoids/therapeutic use , Lupus Erythematosus, Systemic/chemically induced , Lupus Erythematosus, Systemic/drug therapy , Animals , Blotting, Western , Cell Differentiation , Chemokine CXCL1/metabolism , Chemokine CXCL2/metabolism , Disease Models, Animal , Female , Flavonols , Flow Cytometry , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Lupus Erythematosus, Systemic/metabolism , Lupus Nephritis/chemically induced , Lupus Nephritis/drug therapy , Lupus Nephritis/metabolism , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , Terpenes/toxicity , Th17 Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism
19.
Phytomedicine ; 49: 11-22, 2018 Oct 01.
Article in English | MEDLINE | ID: mdl-30217257

ABSTRACT

BACKGROUND: Preterm birth is the most prominent complication attributing to poor pregnancy and neonatal outcome. Infection is most commonly implicated in preterm birth; it initiates a cascade of inflammatory events that leads to the rupture of fetal membranes and spontaneous uterine contractions. Anti-inflammatory agents may thus be a therapeutic approach to prevent the premature rupture of fetal membranes and block contractions. In non-gestational tissues, the polyphenol honokiol has been shown to possess potent anti-inflammatory properties. PURPOSE: The aim of this study was to investigate the effect of honokiol on pro-inflammatory mediators in human gestational tissues. METHODS: Fetal membranes, myometrium and freshly isolated amnion cells and primary myometrial cells were treated with honokiol in the absence or presence of the products lipopolysaccharide (LPS) and fibroblast-stimulating lipopeptide-1 (fsl-1), the viral dsRNA analogue polyinosinic:polycytidylic acid (poly(I:C)) or the pro-inflammatory cytokines TNF or IL1B. A luciferase assay was used to determine the effect of honokiol on nuclear factor kappa B (NF-κB) RelA transcriptional activity. RESULTS: Honokiol significantly decreased pro-inflammatory cytokine (IL1A, IL6) and chemokine (CXCL8, CXCL1, CCL2) mRNA expression and secretion from fetal membranes (amnion and choriodecidua) and myometrium stimulated with LPS, fsl-1 or poly(I:C). In amnion cells, honokiol also significantly decreased the expression and secretion of the extracellular matrix degrading enzyme MMP9. Moreover, in myometrium, honokiol significantly suppressed the expression of the contraction associated protein PTGFR, the secretion of the uterotonic prostaglandins PGE2 and PGF2α, and blocked TNF-induced myometrial cell contractility. Finally, honokiol significantly suppressed IL1B- and TNF-induced NF-κB RelA transcriptional activity in primary amnion and myometrial cells. CONCLUSIONS: Honokiol reduced the expression of pro-inflammatory and pro-labour mediators in human amnion, choriodecidua and myometrium and that this may be facilitated through the suppression of NF-κB activation. These results indicate that the polyphenol honokiol may be a potent therapeutic for the prevention of preterm birth.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Biphenyl Compounds/pharmacology , Extraembryonic Membranes/drug effects , Lignans/pharmacology , Myometrium/drug effects , Chemokine CCL2/metabolism , Chemokine CXCL1/metabolism , Dinoprost/metabolism , Dinoprostone/metabolism , Female , Humans , Interleukin-1beta/pharmacology , Interleukin-8/metabolism , Pregnancy , Premature Birth/prevention & control , Primary Cell Culture , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/pharmacology
20.
Am J Chin Med ; 46(6): 1297-1313, 2018.
Article in English | MEDLINE | ID: mdl-30149752

ABSTRACT

Antrodia cinnamomea, a medicinal mushroom, has previously demonstrated anti-inflammatory activity, although the specific compound responsible for the effect remains unclear. The present study was designed to investigate the anti-inflammatory property of antrolone, a novel benzoid derived from A. cinnamomea mycelium, and to clarify the underlying mechanisms of action. To this end, murine macrophage RAW264.7 cells were treated with antrolone (0.1-30[Formula: see text][Formula: see text]M) 30[Formula: see text]min prior to stimulation with lipopolysaccharides (LPS, 0.1[Formula: see text][Formula: see text]g/ml) for 24[Formula: see text]h. Cell viability, nitric oxide (NO) and prostaglandin E2 (PGE2) production, levels of pro-inflammatory cytokines and chemokines, and the signaling pathways involved in the inflammatory cascades were then investigated. Our results show that antrolone significantly decreased LPS-induced NO, PGE2, pro-inflammatory cytokine, and keratinocyte chemoattractant CXCL1 (KC) production and reduced levels of the proteins inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2). These effects were independent of the effect of antrolone on macrophage cytotoxicity. Moreover, antrolone significantly inhibited the activation of the NF[Formula: see text]B, MAPK, and AKT pathways, while it increased nuclear factor erythroid-2-related factor (Nrf2) and heme oxygenase-1 (HO-1) levels. Our findings suggest that antrolone exhibits potent anti-inflammatory activity and may, therefore, be a lead compound for the development of an anti-inflammatory drug.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antrodia/chemistry , Benzoates/pharmacology , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Animals , Anti-Inflammatory Agents/isolation & purification , Cell Survival/drug effects , Chemokine CXCL1/metabolism , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Dinoprostone/metabolism , Heme Oxygenase-1/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/adverse effects , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , RAW 264.7 Cells , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL