Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Int Immunopharmacol ; 131: 111824, 2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38461633

ABSTRACT

BACKGROUND: Psoriasis is an inflammatory skin disease that occurs repeatedly over time. The natural product of sesquiterpene lactones, Parthenolide (Par), is isolated from Tanacetum parthenium L. (feverfew) which has significant effects on anti-inflammatory. The therapeutic effect of the medication itself is crucial, but different routes of administration of the same drug can also produce different effects. PURPOSE: The aim of our research sought to investigate the ameliorating effects of Par in psoriasis-like skin inflammation and its related mechanism of action. RESULTS: In the IMQ-induced model, intragastric administration of Par reduced the Psoriasis Area and Severity Index (PASI) score, improved skin erythema, scaling, and other symptoms. And Par decreased the expression of Ki67, keratin14, keratin16 and keratin17, and increased the expression of keratin1. Par could reduce IL-36 protein expressions, meanwhile the expression of Il1b, Cxcl1 and Cxcl2 mRNA were also decreased. Par regulated the expression levels of F4/80, MPO and NE. However, skin transdermal administration of Par was more effective. Similarly, Par attenuated IL-36γ, IL-1ß and caspase-1 activated by Poly(I:C) in in vitro and ex vivo. In addition, Par also reduced NE, PR3, and Cathepsin G levels in explant skin tissues. CONCLUSION: Par ameliorated psoriasis-like skin inflammation in both in vivo and in vitro, especially after treatment with transdermal drug delivery, possibly by inhibiting neutrophil extracellular traps and thus by interfering IL-36 signaling pathway. It indicated that Par provides a new research strategy for the treatment of psoriasis-like skin inflammation and is expected to be a promising drug.


Subject(s)
Dermatitis , Extracellular Traps , Psoriasis , Sesquiterpenes , Animals , Mice , Imiquimod/pharmacology , Administration, Cutaneous , Extracellular Traps/metabolism , Skin , Psoriasis/chemically induced , Psoriasis/drug therapy , Psoriasis/metabolism , Sesquiterpenes/therapeutic use , Sesquiterpenes/pharmacology , Dermatitis/drug therapy , Inflammation/chemically induced , Inflammation/drug therapy , Disease Models, Animal , Mice, Inbred BALB C
2.
Food Funct ; 14(5): 2392-2403, 2023 Mar 06.
Article in English | MEDLINE | ID: mdl-36786020

ABSTRACT

Psoriasis is a recurrent inflammatory skin disease. IL-36-related cytokines are overexpressed in psoriasis, but the mechanism is not yet clear. Costunolide (Cos) is a sesquiterpenoid compound derived from the root of the traditional Chinese medicine Aucklandia lappa Decne. This study aimed to explore the mechanism of Cos on improving psoriasis-like skin inflammation. An in vivo model was established by applying imiquimod treatment to the back skin of mice, and an in vitro model was established by using polyinosinic-polycytidylic acid (Poly(I:C)) stimulated-mouse primary dermal fibroblasts to induce inflammation. The results showed that Cos improved the pathological changes of psoriasis-like skin inflammation. In addition, Cos could inhibit epidermal damage and inflammation-related expression and improve the occurrence of skin-related inflammation in both in vivo and in vitro experiments. The improvement of psoriasis-like skin inflammatory response might be through the P2X7R/IL-36 signaling pathway. Collectively, Cos has an inhibitory effect on the expression of psoriasis-like skin inflammation. This showed that Cos has potential skin health promoting benefits by preventing psoriasis-like skin inflammation.


Subject(s)
Dermatitis , Psoriasis , Sesquiterpenes , Animals , Mice , Imiquimod/adverse effects , Skin/metabolism , Psoriasis/chemically induced , Psoriasis/drug therapy , Sesquiterpenes/pharmacology , Sesquiterpenes/therapeutic use , Dermatitis/drug therapy , Dermatitis/etiology , Inflammation/chemically induced , Cytokines/metabolism , Health Promotion , Mice, Inbred BALB C , Disease Models, Animal
3.
J Agric Food Chem ; 70(9): 2968-2983, 2022 Mar 09.
Article in English | MEDLINE | ID: mdl-35212223

ABSTRACT

Digitoflavone (DG) is a natural flavonoid abundant in many fruits, vegetables, and medicinal plants. We investigated whether DG inhibits lipid accumulation and inflammatory responses in alcoholic liver disease (ALD) in vivo and in vitro. The mouse ALD model was established by chronically feeding male C57BL/6 mice an ethanol-containing Lieber-DeCarli liquid diet. In vitro, mouse peritoneal macrophages (MPMs) and mouse bone marrow-derived macrophages (BMDMs) were stimulated with LPS/ATP, whereas HepG2 cells and mouse primary hepatocytes were treated with ethanol. DG reduced the serum levels of transaminase and serum and hepatic levels of triglycerides and malondialdehyde in ALD mice. DG downregulated SREBP1 and its target genes and upregulated PPARα and its target genes in the liver of mice with ALD. DG inhibited TLR4-mediated NLRP3 inflammasome activation, consequently reversing the inflammatory response, including the production of HMGB1, IL-1ß, and IL-36γ, as well as the infiltration of macrophages and neutrophils. DG blocked NLRP3/ASC/caspase-1 inflammasome activation and HMGB1 release in LPS/ATP-stimulated MPMs. When Tlr4 was knocked in LPS/ATP-stimulated BMDMs, HMGB1 production and release were blocked, and NLRP3-mediated cleavage and release of IL-1ß was suppressed in Hmgb1-silenced BMDMs. DG amplified these inhibitory effects in Tlr4 or Hmgb1 knockdown BMDMs. In ethanol-exposed hepatocytes, DG reduced lipogenesis and promoted lipid oxidation by inhibiting the HMGB1-TLR4 signaling pathway while suppressing the inflammatory response induced by ethanol exposure. Our data demonstrated that DG inhibited the occurrence of lipid accumulation and the inflammatory response via the HMGB1-TLR4 axis, underscoring a promising approach and utility of DG for the treatment of ALD.


Subject(s)
Flavones/pharmacology , HMGB1 Protein , Liver Diseases, Alcoholic , Signal Transduction/drug effects , Toll-Like Receptor 4 , Animals , HMGB1 Protein/metabolism , Hep G2 Cells , Humans , Inflammasomes , Liver Diseases, Alcoholic/drug therapy , Liver Diseases, Alcoholic/metabolism , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Toll-Like Receptor 4/metabolism
4.
Food Funct ; 12(21): 10714-10727, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34607339

ABSTRACT

P2X7 receptor (P2X7R) and NLRP3 cooperatively participate in inflammation and hepatocyte damage during hepatic injury induced by lipopolysaccharides (LPS). High-mobility group box 1 (HMGB1) released from immune cells in response to such stimuli plays a vital role in mediating inflammation via TLR4 and the receptor for advanced glycation end products (RAGE), a receptor for HMGB1. However, the correlation among P2X7R, RAGE and TLR4 in regulating the release of HMGB1 has not been elucidated. Increasing the number of daily foods is found to be beneficial for hepatocyte damage in septic hepatic injury. Hence, we investigated the effects of luteolin, a natural flavonoid mainly existing in vegetables and fruits, on liver injury, focusing on how luteolin participates in hepatitis based on the P2X7R-RAGE-TLR4 axis by regulating the release of HMGB1. The results demonstrated that the indicators of hepatic injury such as increased ALT, AST in the serum and infiltration of immune cells were attenuated after luteolin treatment in LPS-induced mice. Luteolin could also suppress the production and release of HMGB1 and the activation of caspase 1 both in LPS-induced mice and LPS/ATP-stimulated HepG2 cells. Collectively, luteolin reversed LPS-induced hepatic injury, especially inflammation, likely by regulating the release of HMGB1 through the P2X7R-RAGE-TLR4 axis.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , HMGB1 Protein/metabolism , Luteolin/pharmacology , Receptors, Purinergic P2X7/metabolism , Sepsis/complications , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Gene Expression Regulation/drug effects , HMGB1 Protein/genetics , Hep G2 Cells , Hepatocytes/drug effects , Humans , Lipopolysaccharides/toxicity , Macrophages, Peritoneal/drug effects , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Receptors, Purinergic P2X7/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
5.
Front Pharmacol ; 12: 723881, 2021.
Article in English | MEDLINE | ID: mdl-34366873

ABSTRACT

Acetaminophen (APAP), one of the most common antipyretic analgesics, which is safe at therapeutic dose, cause acute liver injury and even death at overdose. However, the mechanism of APAP-induced inflammation in liver injury is still controversial. Therefore, effective drug intervention is urgently needed. The aim of this study was to explore the inflammatory exact mechanism of APAP, especially on neutrophils, and to study the intervention effect of Chikusetsusaponin V (CKV) derived from Panax japonicus. Establishment of hepatotoxicity model of APAP in vitro and in vivo. In vitro, HepG2 cells, AML12 cells, primary mouse hepatocytes and neutrophils were used to mimic APAP-affected hepatocytes and neutrophil. In vivo, C57BL/6 mice were administrated overdose of APAP with or without neutrophil depletion or abolishing neutrophil extracellular traps (NETs) formation. In this study, APAP stimulation increased the level of HMGB1, IL-1ß and Caspase-1 in mouse liver, especially hepatocytes, which had a synergistic effect with LPS/ATP combination. NETs were formatted at early stage of APAP or HMGB1-stimulated neutrophils' damage. Conditioned mediums from APAP-treated hepatocytes induced more significant NETs than direct APAP stimulation. Neutrophil depletion or abolishing NETs formation decreased HMGB1 level, eventually blocked hepatocytes necrosis. CKV pretreatment interfered Caspase-1 activation and HMGB1 release in APAP-damaged hepatocytes. CKV also prevented NETs formation. These results indicate that the production of HMGB1 may depend on the activation of Caspase-1 and play a key role in liver inflammation caused by APAP. The cross-dialogue between hepatocytes and neutrophils can be mediated by HMGB1. Therefore, CKV has a positive intervention effect on NETs-related inflammation in APAP-damaged liver, targeting Caspase-1-HMGB1.

6.
Food Funct ; 12(1): 362-372, 2021 Jan 07.
Article in English | MEDLINE | ID: mdl-33325949

ABSTRACT

Excessive alcohol drinking and a high-fat diet (HFD) promote steatohepatitis in the comorbidity of NAFLD and AFLD. Taxifolin (TAX) is a rich dihydroxyflavone compound found in onions, milk thistle and Douglas fir. We aimed to explore the intervention mechanism of TAX on chronic steatohepatitis induced by HFD feeding plus acute ethanol binge. We established an in vivo model by HFD feeding plus a single dose of ethanol binge, and established an in vitro model by oleic acid or palmitic acid on HepG2 cells to induce lipid accumulation. TAX regulated lipid synthesis by inhibiting the expression of SREBP1 and upregulating the PPARγ level. In addition, TAX inhibited the expression of P2X7R, IL-1ß, and caspase-1. Moreover, TAX reduced the expression of caspase-1 activation; thereby inhibiting the recruitment of macrophages and neutrophils. TAX also improved the inflammatory response caused by caspase-1 activation in steatotic hepatocytes. TAX exhibited an inhibitory effect on lipid accumulation and caspase-1-related pyroptosis. Collectively, TAX has therapeutic potential as an intervention of steatohepatitis induced by alcohol combined with HFD and for preventing non-alcoholic fatty liver degeneration targeting caspase-1-dependent pyroptosis.


Subject(s)
Diet, High-Fat/adverse effects , Ethanol/adverse effects , Fatty Liver, Alcoholic/prevention & control , Pyroptosis/drug effects , Quercetin/analogs & derivatives , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Binge Drinking/complications , Cells, Cultured , Central Nervous System Depressants/adverse effects , Disease Models, Animal , Fatty Liver, Alcoholic/etiology , Humans , Male , Mice , Mice, Inbred ICR , Quercetin/pharmacology
7.
Inflammopharmacology ; 28(6): 1481-1493, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33006110

ABSTRACT

Gout, the most prevalent inflammatory arthritis worldwide, released interleukin-1ß (IL-1ß) and Cathepsin B inflammatory mediators that constitute the hallmark of the disease. Herein we aimed to investigate whether procyanidin B2 (PCB2), a natural dietary compound, can suppress MSU crystals-stimulated gouty inflammation. Treated with lipopolysaccharide (LPS) plus MSU, both mouse peritoneal macrophages (MPM) and mouse bone marrow-derived macrophages (BMDM) released a large amount of mature IL-1ß compared to those treated with MSU or LPS alone, while IL-1ß release was blocked by TLR4 and its downstream effector inhibitors. In two mouse models of gout, oral administration of PCB2 suppressed MSU crystals-induced increasing expression of IL-1ß, Cathepsin B and NLRP3 in the air pouch skin and paws, accompanied with the downregulation prostaglandin E2 (PGE2) in pouch exudates. Inflammatory immune cell infiltration including macrophages and neutrophils were significantly blocked by PCB2 in air pouch skin and paws of mice gout groups. PCB2 also suppressed the release of IL-1ß and Cathepsin B induced by MSU plus LPS in MPM. Our results suggest that the inhibitory effects of PCB2 on NLRP3 inflammasome may alleviate inflammatory response in gout, and this might be a promising anti-inflammatory mechanism of PCB2 against the inflammation in gout.


Subject(s)
Biflavonoids/pharmacology , Catechin/pharmacology , Cathepsin B/metabolism , Gout/metabolism , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Proanthocyanidins/pharmacology , Uric Acid/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Dinoprostone/metabolism , Gout/drug therapy , Inflammasomes/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Neutrophils/drug effects , Neutrophils/metabolism
8.
Toxicol Lett ; 333: 22-32, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32721574

ABSTRACT

HSCs (hepatic stellate cells) contribute to the excessive extracellular matrix (ECM) deposition, inflammatory pathways and crucial cell-cell interactions in hepatic disease leading to fibrosis. P2x7R is considered a potential orchestrater in liver fibrosis. For this reason, this work explored the role of P2x7R in liver fibrosis and the mechanism by which P2x7R in macrophages promotes fibrogenesis. In a model of liver fibrosis induced by administration of thioacetamide (TAA), inhibition of P2x7R with its selective inhibitor A438079 reversed TAA-induced liver damage and fibrosis. The mechanism was linked to inhibition of P2x7R-NLRP3 inflammasome activation and thereby infiltration of macrophages and neutrophils into the liver. This result indicated that the P2x7R-TLR4-NLRP3 axis is involved in the process of TGF-ß-mediated ECM deposition in HSCs. Ectopic overexpression of P2x7R lowered the threshold of extracellular matrix (ECM) deposition and maintained HSCs in an activated state. The culture medium of THP-1 macrophages stimulated by LPS/ATP aggravated ECM deposition in HSCs by activating P2x7R. Additionally, IL-1ß secreted by LPS / ATP activated macrophages amplified fibrosis. These data indicate that P2x7R plays a key regulative role in the activation and maintenance of HSCs promoted by macrophages. Thus, pharmacological inhibition of P2x7R could be a potential therapeutic mechanism to treat human liver fibrosis.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Hepatic Stellate Cells/drug effects , Liver Cirrhosis/metabolism , Macrophages, Peritoneal/drug effects , Receptors, Purinergic P2X7/metabolism , Animals , Cell Culture Techniques , Cell Line, Tumor , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Feedback, Physiological , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Liver Cirrhosis/pathology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Inbred C57BL , Purinergic P2X Receptor Antagonists/pharmacology , Pyridines/pharmacology , Tetrazoles/pharmacology , Thioacetamide/toxicity
9.
Biomol Ther (Seoul) ; 28(4): 320-327, 2020 Jul 01.
Article in English | MEDLINE | ID: mdl-32248671

ABSTRACT

In current study, we aimed to investigate whether the gentiopicroside (GPS) derived from Gentiana manshurica Kitagawa could block the progression of alcoholic hepatic steatosis to fibrosis induced by chronic ethanol intake. C57BL/6 mice were fed an ethanol- containing Lieber-DeCarli diet for 4 weeks. LX-2 human hepatic stellate cells were treated with GPS 1 h prior to transforming growth factor-ß (TGF-ß) stimulation, and murine hepatocyte AML12 cells were pretreated by GPS 1 h prior to ethanol treatment. GPS inhibited the expression of type I collagen (collagen I), α-smooth muscle actin (α-SMA) and tissue inhibitor of metal protease 1 in ethanol-fed mouse livers with mild fibrosis. In addition, the imbalanced lipid metabolism induced by chronic ethanol-feeding was ameliorated by GPS pretreatment, characterized by the modulation of lipid accumulation. Consistently, GPS inhibited the expression of collagen I and α-SMA in LX-2 cells stimulated by TGF-ß. Inhibition of lipid synthesis and promotion of oxidation by GPS were also confirmed in ethanol-treated AML12 cells. GPS could prevent hepatic steatosis advancing to the inception of a mild fibrosis caused by chronic alcohol exposure, suggesting GPS might be a promising therapy for targeting the early stage of alcoholic liver disease.

10.
Br J Pharmacol ; 177(12): 2793-2811, 2020 06.
Article in English | MEDLINE | ID: mdl-32022249

ABSTRACT

BACKGROUND AND PURPOSE: Regulating macrophage-hepatocyte crosstalk through P2X7 receptors has led to new pharmacological strategies to reverse alcoholic hepatosteatosis. We investigated how tetrahydroxystilbene glucoside (2354glu), isolated from Polygonum multiflorum, modulates macrophage-hepatocyte crosstalk during alcoholic hepatosteatosis. EXPERIMENTAL APPROACH: A model of alcoholic hepatosteatosis was established by giving ethanol intragastrically to C57BL/6 mice. HepG2 cells were incubated in conditioned medium from LPS+ATP-activated THP-1 human macrophages with silenced or overexpressed P2X7 receptors. THP-1 macrophages or mouse peritoneal macrophages were pretreated with 2354glu for 1 hr prior to LPS+ATP stimulation. Western blots, RT-PCR and immunohistochemical analysis were used, along with over-expression and silencing of P2X7 receptors. KEY RESULTS: Knockdown or overexpression of P2X7 receptors in THP-1 macrophages affected release of mature IL-1ß and, subsequently, modulated lipid metabolism in HepG2 cells via the LKB-AMPK pathway. 2354glu ameliorated alcoholic hepatosteatosis in mice by regulating LKB1-AMPK-SREBP1 pathway and its target genes. Suppression of P2X7 receptor activation by 2354glu inhibited IL-1ß release and reduced macrophage and neutrophil infiltration. In macrophages stimulated with LPS+ATP, expression of P2X7 receptors, caspase-1 and NF-κB, release of IL-1ß, calcium influx and PI uptake were reduced by 2354glu. SIRT1-LKB1-AMPK-SREBP1 axis-mediated lipid accumulation in HepG2 cells was reduced when they were cultured with conditioned media from LPS+ATP-activated THP-1 macrophages pretreated with 2354glu. CONCLUSION AND IMPLICATIONS: Modulation of P2X7 receptors in macrophages regulated lipid accumulation in hepatocytes during alcoholic hepatosteatosis. 2354glu might be a promising candidate that targets P2X7 receptors in macrophages interacting with hepatocytes during alcoholic hepatosteatosis.


Subject(s)
Macrophages , Receptors, Purinergic P2X7 , Adenosine Triphosphate , Animals , Glucosides , Hepatocytes/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Stilbenes
11.
Am J Chin Med ; 47(3): 577-594, 2019.
Article in English | MEDLINE | ID: mdl-30974967

ABSTRACT

Thymoquinone (TQ) is a main aromatic component of Nigella sativa L. seeds or Agastache rugosa (Fisch. & C.A.Mey.) Kuntze. The protective mechanism of TQ against acute liver injury induced by acetaminophen (APAP), however, remains unclear. We aimed to investigated the hepato-protective mechanism of TQ on the development of APAP-induced acute liver injury. Male kunming mice were pretreated with TQ or N-acetylcysteine (NAC) before a single APAP injection. Human Chang liver cells were incubated with TQ, SP600125 or AICAR in presence of APAP for 24 h. TQ pretreatment reduced levels of serum aminotransferases and increased hepatic glutathione and glutathione peroxidase activities via inhibiting CYP2E1 expression. TQ inhibited JNK, ERK and P38 phosphorylation induced by APAP. Meanwhile, TQ inhibited PI3K/mTOR signaling activation and activated AMPK phosphorylation. Moreover, TQ prevented APAP-induced hepatocytes apoptosis regulated by Bcl-2 and Bax. Furthermore, TQ inhibited STAT3 phosphorylation on APAP-induced acute liver injury. In addition, TQ significantly inhibited P2X7R protein expression and IL-1 ß release. APAP-enhanced JNK phosphorylation and APAP-suppressed AMPK phosphorylation were also observed in Chang liver cells, and these changes were recovered by pretreatment with TQ, SP600125 and AICAR. Our findings suggest that TQ may actively prevent APAP-induced acute liver injury, and the effect may be mediated by JNK and AMPK signaling pathways.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Acetaminophen/administration & dosage , Acetaminophen/adverse effects , Benzoquinones/pharmacology , Benzoquinones/therapeutic use , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/prevention & control , Drug Overdose/complications , MAP Kinase Signaling System/drug effects , Phytotherapy , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Cells, Cultured , Chemical and Drug Induced Liver Injury/metabolism , Disease Models, Animal , Hepatocytes/drug effects , Humans , Inflammation , Male , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...