Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.185
Filter
1.
Brain Behav ; 14(7): e3586, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38970230

ABSTRACT

BACKGROUND: Patients with myocardial infarction (MI) frequently experience a heightened incidence of depression, thereby increasing the risk of adverse cardiovascular events. Consequently, early detection and intervention in depressive symptoms among patients with MI are imperative. Shexiang Baoxin Pills (SBP), a Chinese patent medicine employed for the treatment of MI, exhibits diverse mechanisms targeting this condition. Nevertheless, its therapeutic efficacy on postmyocardial infarction depressive symptoms remains unclear. The aim of this study is to investigate the effectiveness and mechanism of SBP in managing depression during acute myocardial infarction (AMI). METHODS: A rat model combining MI and depression was established, and the rats were randomly divided into four groups: the model (MOD) group, SBP group, Fluoxetine (FLX) group, and Sham group. After 28 days of drug intervention, cardiac function was assessed using echocardiography while behavior was evaluated through sucrose preference test (SPT), forced swimming test (FST), and open-field test (OFT). Additionally, levels of inflammatory factors in serum and hippocampus were measured along with NLRP3 inflammasome-related protein expression via Western blotting and immunofluorescence. RESULTS: SBP can enhance cardiac function in rats with AMI and depression, while significantly ameliorating depressive-like behavior. Compared to the Sham group, levels of IL-1ß, IL-18, TNF-α, and other inflammatory factors were markedly elevated in the MOD group. However, expressions of these inflammatory factors were reduced to varying degrees following treatment with SBP or FLX. Analysis of NLRP3 inflammasome-related proteins in the hippocampus revealed a significant upregulation of IL-1ß, IL-18, NLRP3, ASC, caspase-1, and GSDMD in the MOD group; conversely, these measures were significantly attenuated after SBP intervention. CONCLUSION: We have observed a significant amelioration in depression-like behavior upon SBP administration during the treatment of AMI, suggesting that this effect may be attributed to the inhibition of NLRP3-mediated pyroptosis. (The main findings are summarized in the graphical abstract in the supplementary file.).


Subject(s)
Antidepressive Agents , Depression , Drugs, Chinese Herbal , Inflammasomes , Myocardial Infarction , NLR Family, Pyrin Domain-Containing 3 Protein , Rats, Sprague-Dawley , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocardial Infarction/complications , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/administration & dosage , Rats , Depression/drug therapy , Depression/etiology , Antidepressive Agents/pharmacology , Antidepressive Agents/administration & dosage , Male , Inflammasomes/metabolism , Inflammasomes/drug effects , Disease Models, Animal , Signal Transduction/drug effects , Hippocampus/metabolism , Hippocampus/drug effects , Behavior, Animal/drug effects
2.
J Agric Food Chem ; 72(25): 14165-14176, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38872428

ABSTRACT

Atractylodes macrocephala Koidz, a traditional Chinese medicine, contains atractylenolide I (ATR-I), which has potential anticancer, anti-inflammatory, and immune-modulating properties. This study evaluated the therapeutic potential of ATR-I for indomethacin (IND)-induced gastric mucosal lesions and its underlying mechanisms. Noticeable improvements were observed in the histological morphology and ultrastructures of the rat gastric mucosa after ATR-I treatment. There was improved blood flow, a significant decrease in the expression of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), IL-1ß, and IL-18, and a marked increase in prostaglandin E2 (PGE2) expression in ATR-I-treated rats. Furthermore, there was a significant decrease in the mRNA and protein expression levels of NOD-like receptor thermal protein domain associated protein 3 (NLRP3), apoptosis-associated speck-like protein (ASC), cysteinyl aspartate specific proteinase-1 (caspase-1), and nuclear factor-κB (NF-κB) in rats treated with ATR-I. The results show that ATR-I inhibits the NLRP3 inflammasome signaling pathway and effectively alleviates local inflammation, thereby improving the therapeutic outcomes against IND-induced gastric ulcers in rats.


Subject(s)
Atractylodes , Gastric Mucosa , Indomethacin , Inflammasomes , Lactones , NLR Family, Pyrin Domain-Containing 3 Protein , Rats, Sprague-Dawley , Sesquiterpenes , Stomach Ulcer , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Indomethacin/adverse effects , Stomach Ulcer/drug therapy , Stomach Ulcer/chemically induced , Stomach Ulcer/metabolism , Rats , Sesquiterpenes/pharmacology , Sesquiterpenes/chemistry , Lactones/pharmacology , Lactones/chemistry , Inflammasomes/metabolism , Inflammasomes/genetics , Inflammasomes/drug effects , Male , Atractylodes/chemistry , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , NF-kappa B/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/immunology , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-1beta/immunology , Caspase 1/genetics , Caspase 1/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-6/immunology , Interleukin-18/genetics , Interleukin-18/metabolism
3.
J Agric Food Chem ; 72(26): 14701-14712, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38897610

ABSTRACT

Excessive hydrogen peroxide (H2O2) generated during retinal cell metabolic activity could lead to oxidative degeneration of retinal pigment epithelium (RPE) tissue, a specific pathological process implicated in various retinal diseases resulting in blindness, which can be mitigated by taking dietary antioxidants to prevent inflammation and impaired cellular dysfunction. This study tested the hypothesis that damages induced by oxidative stresses can be mitigated by lutein in a H2O2-challenged model, which was based on an ARPE-19 cell monolayer cultured on three-dimensional (3D)-printed fibrous scaffolds. Pretreating these models with lutein (0.5 µM) for 24 h can significantly lower the oxidative stress and maintain phagocytosis and barrier function. Moreover, lutein can modulate the NLRP3 inflammasome, leading to a ∼40% decrease in the pro-inflammatory cytokine (IL-1ß and IL-18) levels. Collectively, this study suggests that the 3D RPE model is an effective tool to examine the capability of lutein to modulate cellular functionalities and regulate NLRP3 inflammation.


Subject(s)
Hydrogen Peroxide , Inflammasomes , Lutein , NLR Family, Pyrin Domain-Containing 3 Protein , Oxidative Stress , Retinal Pigment Epithelium , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/cytology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Humans , Inflammasomes/metabolism , Inflammasomes/drug effects , Hydrogen Peroxide/metabolism , Lutein/pharmacology , Oxidative Stress/drug effects , Cell Line , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Interleukin-18/metabolism , Models, Biological
4.
Int J Nanomedicine ; 19: 4957-4976, 2024.
Article in English | MEDLINE | ID: mdl-38828198

ABSTRACT

Background: The "gut-islets axis" is an important endocrine signaling axis that regulates islets function by modulating the gut microbiota and endocrine metabolism within the gut. However, the specific mechanisms and roles of the intestine in islets regulation remain unclear. Recent studies investigated that exosomes derived from gut microbiota can transport signals to remotely regulate islets ß-cell function, suggesting the possibility of novel signaling pathways mediated by gut exosomes in the regulation of the "gut-islet axis.". Methods: The exosomes were isolated from the intestinal enteroendocrine cell-line STC-1cells culture supernatants treated with palmitate acid (PA) or BSA. Metabolic stress models were established by separately subjecting MIN6 cells to PA stimulation and feeding mice with a high-fat diet. Intervention with exosomes in vitro and in vivo to assess the biological effects of exosomes on islets ß cells under metabolic stress. The Mas receptor antagonist A779 and ACE2ko mice were used to evaluate the role of exosomal ACE2. Results: We found ACE2, a molecule that plays a crucial role in the regulation of islets function, is abundantly expressed in exosomes derived from STC-1 under physiological normal condition (NCEO). These exosomes cannot only be taken up by ß-cells in vitro but also selectively transported to the islets in vivo. Following intervention with NCEXO, both Min6 cells in a lipotoxic environment and mice on a high-fat diet exhibited significant improvements in islets ß-cell function and ß-cell mass. Further investigations demonstrated that these protective effects are attributed to exosomal ACE2, as ACE2 inhibits NLRP3 inflammasome activation and reduces ß-cell pyroptosis. Conclusion: ACE2-enriched exosomes from the gut can selectively target islets, subsequently inhibiting NLRP3 inflammasome activation and ß cell pyroptosis, thereby restoring islets ß cell function under metabolic stress. This study provides novel insights into therapeutic strategies for the prevention and treatment of obesity and diabetes.


Subject(s)
Angiotensin-Converting Enzyme 2 , Exosomes , Inflammasomes , Insulin-Secreting Cells , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Animals , Exosomes/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mice , Pyroptosis/drug effects , Pyroptosis/physiology , Angiotensin-Converting Enzyme 2/metabolism , Inflammasomes/metabolism , Inflammasomes/drug effects , Cell Line , Intestine, Small/drug effects , Male , Diet, High-Fat , Mice, Knockout , Enteroendocrine Cells/drug effects , Enteroendocrine Cells/metabolism
5.
Theranostics ; 14(8): 3082-3103, 2024.
Article in English | MEDLINE | ID: mdl-38855180

ABSTRACT

Background: Gouty arthritis causes severe pain and inflammation. Alginate oligosaccharides (AOSs) are natural products derived from alginate and have anti-inflammatory properties. We explored the potential effects of AOSs with different degrees of polymerization (Dp) on gouty arthritis and associated mechanisms. Methods: We established a mouse model of gouty arthritis by injecting monosodium urate (MSU) into ankle joint. Nocifensive behavior, gait and ankle swelling were used to study AOS's effects. Biochemical assays, in vivo imaging, live cell Ca2+ imaging, electrophysiology, RNA-sequencing, etc. were used for mechanism exploration. Results: AOS2 (Dp=2), AOS3 (Dp=3) and AOS4 (Dp=4) all inhibited ankle swelling, whereas AOS2&3 produced the most obvious analgesia on model mice. AOS3, which was picked for further evaluation, produced dose-dependent ameliorative effects on model mice. AOS3 reversed gait impairments but did not alter locomotor activity. AOS3 inhibited NLRP3 inflammasome activation and inflammatory cytokine up-regulation in ankle joint. AOS3 ameliorated MSU-induced oxidative stress and reactive oxygen species (ROS) production both in vivo and in vitro and reversed the impaired mitochondrial bioenergetics. AOS3 activated the Nrf2 pathway and promoted Nrf2 disassociation from Keap1-bound complex and Nrf2 nuclear translocation, thus facilitating antioxidant gene expression via Nrf2-dependent mechanism. Nrf2 gene deficiency abolished AOS3's ameliorative effects on pain, inflammation and oxidative stress in ankle joints of model mice. AOS3 reduced TRPV1 functional enhancement in DRG neurons and constrained neuroactive peptide release. Conclusions: AOS3 ameliorates gouty arthritis via activating Nrf2-dependent antioxidant signaling, resulting in suppression of ROS-mediated NLRP3 inflammasome activation and TRPV1 enhancement. AOS3 may be novel therapeutics for gouty arthritis.


Subject(s)
Alginates , Arthritis, Gouty , Disease Models, Animal , Inflammation , Oligosaccharides , Animals , Arthritis, Gouty/drug therapy , Arthritis, Gouty/metabolism , Mice , Oligosaccharides/pharmacology , Alginates/pharmacology , Inflammation/drug therapy , Inflammation/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Male , Arthralgia/drug therapy , Arthralgia/metabolism , Uric Acid/metabolism , Mice, Inbred C57BL , Anti-Inflammatory Agents/pharmacology , Inflammasomes/metabolism , Inflammasomes/drug effects , Ankle Joint/pathology , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects
6.
Phytomedicine ; 131: 155758, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38843643

ABSTRACT

BACKGROUND: The adaptor protein apoptosis-associated speck-like protein (ASC) containing a caspase recruitment domain (CARD) can be activated through pyrin domain (PYD) interactions between sensors and ASC, and through CARD interactions between caspase-1 and ASC. Although the majority of ternary inflammasome complexes depend on ASC, drugs targeting ASC protein remain scarce. After screening natural compounds from Isatidis Radixin, we found that tryptanthrin (TPR) could inhibit NLRP3-induced IL-1ß and caspase-1 production, but the underlying anti-inflammatory mechanisms remain to be elucidated. PURPOSE: The purpose of this study was to determine the impact of TPR on the NLRP3, NLRC4, and AIM2 inflammasomes and the underlying mechanisms. Additionally, the efficacy of TPR was analysed in the further course of methionine- and choline-deficient (MCD)-induced NASH and lipopolysaccharide (LPS)-induced sepsis models of mice. METHODS: In vitro studies used bone marrow-derived macrophages to assess the anti-inflammatory activity of TPR, and the techniques included western blot, testing of intracellular K+ and Ca2+, immunofluorescence, enzyme-linked immunosorbent assay (ELISA), co-immunoprecipitation, ASC oligomerization assay, surface plasmon resonance (SPR), and molecular docking. We used LPS-induced sepsis models and MCD-induced NASH models in vivo to evaluate the effectiveness of TPR in inhibiting inflammatory diseases. RESULTS: Our observations suggested that TPR could inhibit NLRP3, NLRC4, and AIM2 inflammasome activation. As shown in a mouse model of inflammatory diseases caused by MCD-induced NASH and LPS-induced sepsis, TPR significantly alleviated the progression of diseases. TPR interrupted the interactions between ASC and NLRP3/NLRC4/AIM2 in the co-immunoprecipitation experiment, and stable binding of TPR to ASC was also evident in SPR experiments. The underlying mechanisms of anti-inflammatory activities of TPR might be associated with targeting ASC, in particular, PYD domain of ASC. CONCLUSION: In general, the requirement for ASC in multiple inflammasome complexes makes TPR, as a novel broad-spectrum inflammasome inhibitor, potentially useful for treating a wide range of multifactorial inflammasome-related diseases.


Subject(s)
CARD Signaling Adaptor Proteins , Calcium-Binding Proteins , Inflammasomes , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Non-alcoholic Fatty Liver Disease , Quinazolines , Animals , Inflammasomes/metabolism , Inflammasomes/drug effects , CARD Signaling Adaptor Proteins/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Male , Calcium-Binding Proteins/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Quinazolines/pharmacology , Mice , Apoptosis Regulatory Proteins/metabolism , Interleukin-1beta/metabolism , DNA-Binding Proteins/metabolism , Caspase 1/metabolism , Sepsis/drug therapy , Anti-Inflammatory Agents/pharmacology , Lipopolysaccharides , Macrophages/drug effects , Macrophages/metabolism , Disease Models, Animal
7.
Phytomedicine ; 131: 155771, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38851101

ABSTRACT

BACKGROUND: Sepsis often leads to significant morbidity and mortality due to severe myocardial injury. As is known, the activation of NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome crucially contributes to septic cardiomyopathy (SCM) by facilitating the secretion of interleukin (IL)-1ß and IL-18. The removal of palmitoyl groups from NLRP3 is a crucial step in the activation of the NLRP3 inflammasome. Thus, the potential inhibitors that regulate the palmitoylation and inactivation of NLRP3 may significantly diminish sepsis-induced cardiac dysfunction. PURPOSE: The present study sought to explore the effects of the prospective flavonoid compounds targeting NLRP3 on SCM and to elucidate the associated underlying mechanisms. STUDY DESIGN: The palmitoylation and activation of NLRP3 were detected in H9c2 cells and C57BL/6 J mice. METHODS/RESULTS: Echocardiography, histological staining, western blotting, co-immunoprecipitation, qPCR, ELISA and network pharmacology were used to assess the impact of vaccarin (VAC) on SCM in mice subjected to lipopolysaccharide (LPS) injection. From the collection of 74 compounds, we identified that VAC had the strongest capability to suppress NLRP3 luciferase report gene activity in cardiomyocytes, and the anti-inflammatory characteristics of VAC were further ascertained by the network pharmacology. Exposure of LPS triggered apoptosis, inflammation, oxidative stress, mitochondrial disorder in cardiomyocytes. The detrimental alterations were significantly reversed upon VAC treatment in both septic mice and H9c2 cells exposed to LPS. In vivo experiments demonstrated that VAC treatment alleviated septic myocardial injury, indicated by enhanced cardiac function parameters, preserved cardiac structure, and reduced inflammation/oxidative response. Mechanistically, VAC induced NLRP3 palmitoylation to inactivate NLRP3 inflammasome by acting on zDHHC12. In support, the NLRP3 agonist ATP and the acylation inhibitor 2-bromopalmitate (2-BP) prevented the effects of VAC. CONCLUSION: Our findings suggest that VAC holds promise in protecting against SCM by mitigating cardiac oxidative stress and inflammation via priming NLRP3 palmitoylation and inactivation. These results lay the solid basis for further assessment of the therapeutic potential of VAC against SCM.


Subject(s)
Cardiomyopathies , Inflammasomes , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Sepsis , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Cardiomyopathies/drug therapy , Sepsis/drug therapy , Sepsis/complications , Mice , Male , Inflammasomes/metabolism , Inflammasomes/drug effects , Lipoylation/drug effects , Rats , Oxidative Stress/drug effects , Cell Line , Lipopolysaccharides , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Interleukin-1beta/metabolism , Interleukin-18/metabolism
8.
Phytomedicine ; 131: 155765, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38851105

ABSTRACT

BACKGROUND: Infection by Toxoplasma gondii can lead to severe pneumonia, with current treatments being highly inadequate. The NLRP3 inflammasome is one member of the NOD-like receptor family with a pyrin domain, which is crucial in the innate immune defense against T. gondii. Research has shown that resveratrol (RSV) prevents lung damage caused by this infection by inhibiting the T. gondii-derived heat shock protein 70/TLR4/NF-κB pathway, thus reducing the macrophage-driven inflammatory response. However, it should be mentioned that the participation of NLRP3 inflammasome in the immune response to the lung injuries caused by T. gondii infections is not entirely clear. PURPOSE: This study aims to clarify how RSV ameliorates lung damage triggered by Toxoplasma gondii infection, with a particular focus on the pathway involving TLR4, NF-κB, and the NLRP3 inflammasome. METHODS: Both in vitro and in vivo models of infection were developed by employing the RH strain of T. gondii in BALB/c mice and RAW 264.7 macrophage cell lines. The action mechanism of RSV was explored using techniques such as molecular docking, surface plasmon resonance, ELISA, Western blot, co-immunoprecipitation, and immunofluorescence staining. RESULTS: Findings indicate that the suppression of TLR4 or NF-κB impacts the levels of proteins associated with the NLRP3 inflammasome pathway. Additionally, a significant affinity for binding between RSV and NLRP3 was observed. Treatment with RSV led to a marked reduction in the activation and formation of the NLRP3 inflammasome within lung tissues and RAW 264.7 cells, alongside a decrease in IL-1ß concentrations in the bronchoalveolar lavage fluid. These outcomes align with those seen when using the NLRP3 inhibitor CY-09. Moreover, the application of CY-09 prior to RSV negated the latter's anti-inflammatory properties. CONCLUSION: Considering insights from previous research alongside the outcomes of the current investigation, it appears that the TLR4/NF-κB/NLRP3 signaling pathway emerges as a promising target for immunomodulation to alleviate lung injury from T. gondii infection. The evidence gathered in this study lays the groundwork for the continued exploration and potential future clinical deployment of RSV as a therapeutic agent with anti-Toxoplasma properties and the capability to modulate the inflammatory response.


Subject(s)
Inflammasomes , Mice, Inbred BALB C , NF-kappa B , NLR Family, Pyrin Domain-Containing 3 Protein , Pneumonia , Resveratrol , Toll-Like Receptor 4 , Toxoplasma , Resveratrol/pharmacology , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mice , Inflammasomes/drug effects , Inflammasomes/metabolism , RAW 264.7 Cells , Toll-Like Receptor 4/metabolism , Pneumonia/drug therapy , Pneumonia/parasitology , Toxoplasma/drug effects , NF-kappa B/metabolism , Toxoplasmosis/drug therapy , Lung/drug effects , Lung/parasitology , Molecular Docking Simulation , Female , Signal Transduction/drug effects , Macrophages/drug effects
9.
J Physiol Investig ; 67(3): 139-152, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38902958

ABSTRACT

Inflammatory bowel disease (IBD) comprises a group of idiopathic intestinal disorders, including ulcerative colitis and Crohn's disease, significantly impacting the quality of life for affected individuals. The effective management of these conditions remains a persistent challenge. The NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, a complex molecular structure, regulates the production of pro-inflammatory cytokines such as interleukin-1ß. Abnormal activation of the NLRP3 inflammasome plays a pivotal role in the development of IBD, making it a compelling target for therapeutic intervention. Our research revealed that cinnamaldehyde (CA), a major bioactive compound found in the leaves of Cinnamomum osmophloeum kaneh, demonstrated a remarkable ability to alleviate colitis induced by dextran sulfate sodium (DSS) in a mouse model. This effect was attributed to CA's ability to downregulate the activation of the NLRP3 inflammasome and reduce the expression of pro-inflammatory mediators in the colon. In the mechanism study, we observed that CA inhibited the NLRP3 inflammasome in macrophages, at least partially, by enhancing the autophagic response, without reducing mitochondrial damage. These findings collectively suggest that CA holds significant potential as a therapeutic agent for enhancing the management of IBD, offering a promising avenue for further research and development.


Subject(s)
Acrolein , Cinnamomum , Colitis , Dextran Sulfate , Inflammasomes , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Plant Leaves , Animals , Acrolein/analogs & derivatives , Acrolein/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Mice , Colitis/chemically induced , Colitis/drug therapy , Cinnamomum/chemistry , Inflammasomes/drug effects , Inflammasomes/metabolism , Plant Leaves/chemistry , Male
10.
J Physiol Investig ; 67(3): 129-138, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38902960

ABSTRACT

Ischemia-reperfusion (IR) injury remains a pivotal contributor to myocardial damage following acute coronary events and revascularization procedures. Phosphoinositide 3-kinase (PI3K), a key mediator of cell survival signaling, plays a central role in regulating inflammatory responses and cell death mechanisms. Trans-chalcone (Tch), a natural compound known for its anti-inflammatory activities, has shown promise in various disease models. The aim of the current study was to investigate the potential protective effects of Tch against myocardial injury induced by ischemia and reperfusion challenges by targeting the PI3K-inflammasome interaction. Experimental models utilizing male rats subjected to an in vivo model of IR injury and myocardial infarction were employed. Administration of Tch (100 µg/kg, intraperitoneally) significantly reduced myocardial injury, as indicated by limited infarct size and decreased levels of the myocardial enzyme troponin. Mechanistically, Tch upregulated PI3K expression, thereby inhibiting the activity of the NOD-like receptor protein 3 inflammasome followed by the activation of pro-inflammatory cytokines interleukin-1ß (IL-1ß) and IL-18. Moreover, it mitigated oxidative stress and suppressed vascular-intercellular adhesion molecules, contributing to its cardioprotective effects. The PI3K/Akt pathway inhibitor LY294002 considerably attenuated the beneficial effects of Tch. These findings highlight the therapeutic potential of Tch in ameliorating myocardial injury associated with IR insults through its modulation of the PI3K/Akt-inflammasome axis. The multifaceted mechanisms underlying its protective effects signify Tch as a promising candidate for further exploration in developing targeted therapies aimed at mitigating ischemic heart injury and improving clinical outcomes in cardiovascular diseases characterized by IR injury.


Subject(s)
Myocardial Reperfusion Injury , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Rats, Sprague-Dawley , Animals , Male , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/pathology , Proto-Oncogene Proteins c-akt/metabolism , Rats , Phosphatidylinositol 3-Kinases/metabolism , Inflammasomes/metabolism , Inflammasomes/drug effects , Chalcone/pharmacology , Chalcone/analogs & derivatives , Chalcone/therapeutic use , Signal Transduction/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
11.
BMC Complement Med Ther ; 24(1): 213, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38835032

ABSTRACT

BACKGROUND: Gastrodin (GAS), a main bioactive component of the herbal plant, Gastrodia elata Blume, has shown to have beneficial effects on neuroinflammatory diseases such as Alzheimer's disease in animal studies and migraine in clinical studies. Inflammasome is a multimeric protein complex having a core of pattern recognition receptor and has been implicated in the development of neuroinflammatory diseases. Gastrodin has shown to modulate the activation of nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome. This study investigated the effects of GAS on the intensity of mechanical allodynia and associated changes in NLRP3 inflammasome expression at the spinal level using L5/6 spinal nerve ligation model (SNL) in rats. METHODS: Intrathecal (IT) catheter implantation and SNL were used for drug administration and pain model in male Sprague-Dawley rats. The effect of gastrodin or MCC950 (NLRP3 inflammasome inhibitor) on mechanical allodynia was measured by von Frey test. Changes in NLRP3 inflammasome components and interleukin-1ß (IL-1ß) and cellular expression were examined in the spinal cord and dorsal root ganglion. RESULTS: The expression of NLRP3 inflammasome components was found mostly in the neurons in the spinal cord and dorsal root ganglion. The protein and mRNA levels of NLRP3, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), caspase-1, and IL-1ß were upregulated in SNL animals compared to Sham animals. IT administration of GAS significantly attenuated the expression of NLRP3 inflammasome and the intensity of SNL-induced mechanical allodynia. NLRP3 inflammasome inhibitor, MCC950, also attenuated the intensity of allodynia, but the effect is less strong and shorter than that of GAS. CONCLUSIONS: Expression of NLRP3 inflammasome and IL-1ß is greatly increased and mostly found in the neurons at the spinal level in SNL model, and IT gastrodin exerts a significant anti-allodynic effect in SNL model partly through suppressing the expression of NLRP3 inflammasome.


Subject(s)
Benzyl Alcohols , Disease Models, Animal , Glucosides , Hyperalgesia , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Rats, Sprague-Dawley , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Benzyl Alcohols/pharmacology , Glucosides/pharmacology , Male , Rats , Inflammasomes/metabolism , Inflammasomes/drug effects , Hyperalgesia/drug therapy , Spinal Nerves/drug effects , Injections, Spinal
12.
Chin J Nat Med ; 22(6): 486-500, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38906597

ABSTRACT

Neuroinflammation, mediated by the nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing-3 (NLRP3) inflammasome, is a significant contributor to the pathogenesis of neurodegenerative diseases (NDDs). Reynosin, a natural sesquiterpene lactone (SL), exhibits a broad spectrum of pharmacological effects, suggesting its potential therapeutic value. However, the effects and mechanism of reynosin on neuroinflammation remain elusive. The current study explores the effects and mechanisms of reynosin on neuroinflammation using mice and BV-2 microglial cells treated with lipopolysaccharide (LPS). Our findings reveal that reynosin effectively reduces microglial inflammation in vitro, as demonstrated by decreased CD11b expression and lowered interleukin-1 beta (IL-1ß) and interleukin-18 (IL-18) mRNA and protein levels. Correspondingly, in vivo, results showed a reduction in the number of Iba-1 positive cells and alleviation of morphological alterations, alongside decreased expressions of IL-1ß and IL-18. Further analysis indicates that reynosin inhibits NLRP3 inflammasome activation, evidenced by reduced transcription of NLRP3 and caspase-1, diminished NLRP3 protein expression, inhibited apoptosis-associated speck-like protein containing a CARD (ASC) oligomerization, and decreased caspase-1 self-cleavage. Additionally, reynosin curtailed the activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, demonstrated by reduced NADP+ and NADPH levels, downregulation of gp91phox mRNA, protein expression, suppression of p47phox expression and translocation to the membrane. Moreover, reynosin exhibited a neuroprotective effect against microglial inflammation in vivo and in vitro. These collective findings underscore reynosin's capacity to mitigate microglial inflammation by inhibiting the NLRP3 inflammasome, thus highlighting its potential as a therapeutic agent for managing neuroinflammation.


Subject(s)
Inflammasomes , Microglia , NADPH Oxidases , NLR Family, Pyrin Domain-Containing 3 Protein , Sesquiterpenes , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Microglia/drug effects , Microglia/metabolism , Mice , Inflammasomes/metabolism , Inflammasomes/drug effects , Sesquiterpenes/pharmacology , NADPH Oxidases/metabolism , Neurons/drug effects , Neurons/metabolism , Mice, Inbred C57BL , Neuroinflammatory Diseases/drug therapy , Male , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Lipopolysaccharides , Interleukin-18/metabolism , Cell Line , Inflammation/drug therapy , Inflammation/metabolism
13.
Chin J Nat Med ; 22(6): 515-529, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38906599

ABSTRACT

Depression ranks among the most common neuropsychiatric disorders globally. Current studies examining the roles of inflammation and mitochondrial autophagy in the antidepressant efficacy of paeoniflorin (PF) are sparse. This study aimed to elucidate PF's antidepressant mechanism by promoting autophagy and inhibiting NLRP3 inflammasome activation using chronic unpredictable mild stimulation (CUMS)-induced C57BL/6 mouse models in vivo and corticosterone (CORT)-induced HT22 cell models in vitro. Results demonstrated that PF enhanced the viability of HT22 cells following CORT exposure, restored mitochondrial membrane potential (MMP), reduced reactive oxygen species accumulation, increased LC3 fluorescence intensity, and suppressed inflammatory cytokine secretion and inflammation activation. Additionally, PF ameliorated depressive behaviors induced by CUMS and improved damage in hippocampal neurons. It also reduced the expression of NLRP3, ASC, Caspase-1, IL-1ß, and the assembly of the NLRP3 inflammasome. Moreover, PF upregulated the expression of autophagy-related proteins in the hippocampus, facilitating the clearance of damaged mitochondria and enhancing autophagy. The role of autophagy in PF's antidepressant effects was further confirmed through the use of the autophagy inhibitor 3-methyladenine (3-MA), which reduced the efficacy of PF. In conclusion, PF effectively improved depressive behaviors in CUMS-induced mice and reduced NLRP3-mediated inflammation both in vivo and in vitro, likely via the induction of autophagy.


Subject(s)
Autophagy , Depression , Glucosides , Inflammasomes , Mice, Inbred C57BL , Mitochondria , Monoterpenes , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Glucosides/pharmacology , Autophagy/drug effects , Monoterpenes/pharmacology , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Inflammasomes/metabolism , Inflammasomes/drug effects , Male , Depression/drug therapy , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Hippocampus/drug effects , Hippocampus/metabolism , Disease Models, Animal , Reactive Oxygen Species/metabolism
14.
Molecules ; 29(12)2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38930963

ABSTRACT

Ulcerative colitis (UC) is difficult to cure and easy to relapse, leading to poor quality of life for patients. Oxymatrine (OMT) is one of the main alkaloids of Sophora flavescens Aiton, which has many effects, such as anti-inflammation, anti-oxidative stress, and immunosuppression. This study aimed to investigate whether OMT could attenuate ulcerative colitis by inhibiting the NOD-like receptor family pyrin domain containing three (NLRP3) inflammasome-mediated pyroptosis. In this study, the UC rat models were established by 2,4,6-Trinitrobenzenesulfonic acid (TNBS) in vivo, while RAW264.7 cells and peritoneal macrophages were stimulated with Lipopolysaccharides/Adenosine Triphosphate (LPS/ATP) in vitro to simulate pyroptosis models, and Western blotting (WB) and other detection techniques were applied to analyze proteins involved in the NLRP3 inflammasome pathway. Our results showed that OMT alleviated colitis ulcers and pathological damage in the TNBS-induced UC rats and exhibited an inhibitory effect on pyroptosis at the early stage of UC. In the model group, the pyroptosis reached the peak at 24 h after modeling with the contents of active-cysteine-aspartic proteases-1 (caspase-1), Gasdermin D (GSDMD)-N, and cleaved-interleukin-1 beta (IL-1ß) to the highest expression level. Meanwhile, we found that OMT (80 mg kg-1) remarkably decreased the expression levels of NLRP3, active-caspase-1, and cleaved-IL-1ß at 24 h in the lesion tissue from UC rats. Further experiments on cells demonstrated that OMT at concentrations of 100 and 250 µM significantly inhibited cell death caused by NLRP3 inflammasome activation (p < 0.05), downregulated caspase-1, GSDMD, and decreased the levels of active-caspase-1, GSDMD-N, cleaved-IL-1ß in RAW326.7 cells, and peritoneal macrophages. In summary, these results indicated that OMT could attenuate ulcerative colitis through inhibiting pyroptosis mediated by the NLRP3 inflammasome. The inhibition of the NLRP3 inflammasome may be a potential strategy for UC.


Subject(s)
Alkaloids , Colitis, Ulcerative , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Quinolizines , Animals , Quinolizines/pharmacology , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Alkaloids/pharmacology , Pyroptosis/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mice , Rats , Inflammasomes/metabolism , Inflammasomes/drug effects , RAW 264.7 Cells , Male , Disease Models, Animal , Rats, Sprague-Dawley , Trinitrobenzenesulfonic Acid , Lipopolysaccharides , Matrines
15.
Biosci Biotechnol Biochem ; 88(7): 759-767, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38719485

ABSTRACT

Our previous studies have demonstrated that Mito-Tempol (also known as 4-hydroxy-Tempo), a mitochondrial reactive oxygen species scavenger, alleviates oxidized low-density lipoprotein (ox-LDL)-triggered foam cell formation. Given the effect of oxidative stress on activating the NOD-, LRR-, and pyrin domain-containing 3 (NLRP3) inflammasome, which promotes foam cell formation, we aimed to explore whether Mito-Tempo inhibits ox-LDL-triggered foam cell formation by regulating NLRP3 inflammasome. The results revealed that Mito-Tempo re-activated Nrf2 and alleviated macrophage foam cell formation induced by ox-LDL, whereas the effects were reversed by ML385 (a specific Nrf2 inhibitor). Mito-Tempo restored the expression and nuclear translocation of Nrf2 by decreasing ox-LDL-induced ubiquitination. Furthermore, Mito-Tempo suppressed ox-LDL-triggered NLRP3 inflammasome activation and subsequent pyroptosis, whereas the changes were blocked by ML385. Mito-Tempo decreased lipoprotein uptake by inhibiting CD36 expression and suppressed foam cell formation by regulating the NLRP3 inflammasome. Taken together, Mito-Tempo exhibits potent anti-atherosclerotic effects by regulating Nrf2/NLRP3 signaling.


Subject(s)
Foam Cells , Lipoproteins, LDL , NF-E2-Related Factor 2 , NLR Family, Pyrin Domain-Containing 3 Protein , Signal Transduction , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Lipoproteins, LDL/metabolism , NF-E2-Related Factor 2/metabolism , Foam Cells/drug effects , Foam Cells/metabolism , Signal Transduction/drug effects , Mice , Animals , Inflammasomes/metabolism , Inflammasomes/drug effects , Pyroptosis/drug effects , Humans , RAW 264.7 Cells , Cyclic N-Oxides/pharmacology , CD36 Antigens/metabolism , Organophosphorus Compounds , Piperidines
16.
Phytomedicine ; 130: 155693, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38763006

ABSTRACT

BACKGROUND: Alcoholic liver disease (ALD), a public health challenge worldwide caused by long-term persistent drinking, is life-threatening with minimal approved therapies. Hepatic steatosis accompanied by inflammation is an initial and inevitable stage in the complex progression of simple alcoholic liver injury to more severe liver diseases such as hepatitis, liver fibrosis, cirrhosis and liver cancer. PURPOSE: We aimed to identify the therapeutic role of Bruceine A (BA) in ALD whilst attempting to explore whether its protective effects depend specifically on the farnesoid X receptor (FXR). METHODS: Autodock was applied to detect the affinity between BA and FXR. Lieber-DeCarli liquid diet with 5 % ethanol (v/v) was adopted to establish the mouse ALD model. The lentivirus mediating FXR (LV-FXR) was injected into mice via the tail vein to establish FXR-overexpressed mice. FXR silencing or overexpression plasmids were transfected into AML-12 cells prior to ethanol stimulation. Quantitative real-time PCR, Western blotting and immunofluorescence assays were employed to determine the expression of related genes. We subjected liver sections to H&E and Oil Red O staining to evaluate the liver histological injury and the deposition of lipid droplets. RESULTS: BA significantly reduced body weight and liver-to-body weight ratios as well as biochemical indexes in mice. Ethanol-induced liver damage and lipid accumulation could be alleviated by BA treatment. BA bound to FXR by two hydrogen bonds. There was a positive correlation between BA administration and FXR expression. BA inhibited the expression of lipid synthesis genes and enhanced the expression of lipid metabolism genes by activating FXR, thus alleviating steatosis in ALD. Moreover, BA exerted an ameliorative effect against inflammation by inhibiting the activation of absent in melanoma 2 (AIM2) inflammasome by activating FXR. FXR overexpression possessed the ability to counter the accumulation of lipid and the activation of AIM2 inflammasome caused by ethanol. FXR deficiency exacerbated ethanol-induced liver steatosis and inflammation. The hepatoprotective effect of BA could be disrupted by FXR antagonist guggulsterone (GS) in vivo and FXR siRNA in vitro. CONCLUSION: BA alleviated alcoholic liver disease by inhibiting AIM2 inflammasome activation through an FXR-dependent mechanism. This study may potentially represent a new therapeutic approach for ALD.


Subject(s)
Inflammasomes , Liver Diseases, Alcoholic , Mice, Inbred C57BL , Receptors, Cytoplasmic and Nuclear , Animals , Receptors, Cytoplasmic and Nuclear/metabolism , Liver Diseases, Alcoholic/drug therapy , Inflammasomes/metabolism , Inflammasomes/drug effects , Male , Mice , Disease Models, Animal , Liver/drug effects , Liver/pathology , Liver/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Ethanol
17.
Nanomedicine ; 59: 102752, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38740358

ABSTRACT

Fisetin has displayed potential as an anticonvulsant in preclinical studies yet lacks clinical validation. Challenges like low solubility and rapid metabolism may limit its efficacy. This study explores fisetin-loaded chitosan nanoparticles (NP) to address these issues. Using a murine model of pilocarpine-induced temporal lobe epilepsy, we evaluated the anticonvulsant and neuroprotective effects of fisetin NP. Pilocarpine-induced seizures and associated neurobehavioral deficits were assessed after administering subtherapeutic doses of free fisetin and fisetin NP. Changes in ROS, inflammatory cytokines, and NLRP3/IL-18 expression in different brain regions were estimated. The results demonstrate that the fisetin NP exerts protection against seizures and associated depression-like behavior and memory impairment. Furthermore, biochemical, and histological examinations supported behavioral findings suggesting attenuation of ROS/TNF-α-NLRP3 inflammasome pathway as a neuroprotective mechanism of fisetin NP. These findings highlight the improved pharmacodynamics of fisetin using fisetin NP against epilepsy, suggesting a promising therapeutic approach against epilepsy and associated behavioral deficits.


Subject(s)
Chitosan , Epilepsy, Temporal Lobe , Flavonols , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Nanoparticles , Pilocarpine , Reactive Oxygen Species , Tumor Necrosis Factor-alpha , Animals , Epilepsy, Temporal Lobe/drug therapy , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/pathology , Epilepsy, Temporal Lobe/metabolism , Chitosan/chemistry , Chitosan/pharmacology , Flavonols/pharmacology , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Inflammasomes/drug effects , Nanoparticles/chemistry , Male , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism , Flavonoids/pharmacology , Flavonoids/administration & dosage , Behavior, Animal/drug effects , Anticonvulsants/pharmacology , Neuroprotective Agents/pharmacology
18.
Phytomedicine ; 129: 155703, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38723527

ABSTRACT

BACKGROUND: Non-alcoholic steatohepatitis (NASH), the inflammatory subtype in the progression of non-alcoholic fatty liver disease, is becoming a serious burden threatening human health, but no approved medication is available to date. Mononoside is a natural active substance derived from Cornus officinalis and has been confirmed to have great potential in regulating lipid metabolism in our previous studies. However, its effect and mechanism to inhibit the progression of NASH remains unclear. PURPOSE: Our work aimed to explore the action of mononoside in delaying the progression of NASH and its regulatory mechanisms from the perspective of regulating lipophagy. METHODS AND RESULTS: Male C57BL/6 mice were fed with a high-fat and high-fructose diet for 16 weeks to establish a NASH mouse model. After 8 weeks of high-fat and high-fructose feeding, these mice were administrated with different doses of morroniside. H&E staining, ORO staining, Masson staining, RNA-seq, immunoblotting, and immunofluorescence were performed to determine the effects and molecular mechanisms of morroniside in delaying the progression of NASH. In this study, we found that morroniside is effective in attenuating hepatic lipid metabolism disorders and inflammatory response activation, thereby limiting the progression from simple fatty liver to NASH in high-fat and high-fructose diet-fed mice. Mechanistically, we identified AMPK signaling as the key molecular pathway for the positive efficacy of morroniside by transcriptome sequencing. Our results revealed that morroniside maintained hepatic lipid metabolism homeostasis and inhibited NLRP3 inflammasome activation by promoting AMPKα phosphorylation-mediated lipophagy and fatty acid oxidation. Consistent results were observed in palmitic acid-treated cell models. Of particular note, silencing AMPKα both in vivo and in vitro reversed morroniside-induced lipophagy flux enhancement and NLRP3 inflammasome inhibition, emphasizing the critical role of AMPKα activation in the effect of morroniside in inhibiting NASH progression. CONCLUSION: In summary, the present study provides strong evidence for the first time that morroniside inhibits NASH progression by promoting AMPK-dependent lipophagy and inhibiting NLRP3 inflammasome activation, suggesting that morroniside is expected to be a potential molecular entity for the development of therapeutic drugs for NASH.


Subject(s)
AMP-Activated Protein Kinases , Diet, High-Fat , Lipid Metabolism , Non-alcoholic Fatty Liver Disease , Animals , Humans , Male , Mice , AMP-Activated Protein Kinases/metabolism , Cornus/chemistry , Diet, High-Fat/adverse effects , Disease Models, Animal , Disease Progression , Fructose , Glycosides/pharmacology , Inflammasomes/metabolism , Inflammasomes/drug effects , Lipid Metabolism/drug effects , Liver/drug effects , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy
19.
Biomed Pharmacother ; 175: 116649, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38692059

ABSTRACT

BACKGROUND: Second-generation antipsychotics increase the risk of atrial fibrillation. This study explores whether the atypical antipsychotic ziprasidone triggers inflammasome signaling, leading to atrial arrhythmia. METHODS: Electromechanical and pharmacological assessments were conducted on the rabbit left atria (LA). The patch-clamp technique was used to measure ionic channel currents in single cardiomyocytes. Detection of cytosolic reactive oxygen species production was performed in atrial cardiomyocytes. RESULTS: The duration of action potentials at 50 % and 90 % repolarization was dose-dependently shortened in ziprasidone-treated LA. Diastolic tension in LA increased after ziprasidone treatment. Ziprasidone-treated LA showed rapid atrial pacing (RAP) triggered activity. PI3K inhibitor, Akt inhibitor and mTOR inhibitor abolished the triggered activity elicited by ziprasidone in LA. The NLRP3 inhibitor MCC950 suppressed the ziprasidone-induced post-RAP-triggered activity. MCC950 treatment reduced the reverse-mode Na+/Ca2+ exchanger current in ziprasidone-treated myocytes. Cytosolic reactive oxygen species production decreased in ziprasidone-treated atrial myocytes after MCC950 treatment. Protein levels of inflammasomes and proinflammatory cytokines, including NLRP3, caspase-1, IL-1ß, IL-18, and IL-6 were observed to be upregulated in myocytes treated with ziprasidone. CONCLUSIONS: Our findings suggest ziprasidone induces atrial arrhythmia, potentially through upregulation of the NLRP3 inflammasome and enhancement of reactive oxygen species production via the PI3K/Akt/mTOR pathway.


Subject(s)
Atrial Fibrillation , Inflammasomes , Myocytes, Cardiac , Piperazines , Proto-Oncogene Proteins c-akt , Reactive Oxygen Species , Signal Transduction , TOR Serine-Threonine Kinases , Animals , Atrial Fibrillation/chemically induced , Atrial Fibrillation/metabolism , TOR Serine-Threonine Kinases/metabolism , Inflammasomes/metabolism , Inflammasomes/drug effects , Signal Transduction/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rabbits , Reactive Oxygen Species/metabolism , Piperazines/pharmacology , Male , Phosphatidylinositol 3-Kinases/metabolism , Thiazoles/pharmacology , Heart Atria/drug effects , Heart Atria/metabolism , Action Potentials/drug effects , Antipsychotic Agents/pharmacology
20.
Phytomedicine ; 130: 155556, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38810552

ABSTRACT

BACKGROUND: Systemic lupus erythematosus (SLE) is a multi-system autoimmune disease that affects multiple organs and cause a wide range of severe clinical manifestations, including lupus nephritis (LN), which is a major risk factor for morbidity and mortality in individual with SLE. Ursolic acid (UA) is a natural compound with favorable anti-inflammatory properties and has been employed to treat multiple disease, including inflammatory diseases, diabetes, and Parkinson's disease. However, its therapeutic potential on LN and the underlying mechanisms remains unclear. PURPOSE: This aim of this study was to investigate the impact of UA on LN and its underlying mechanism. METHODS: MRL/lpr lupus-prone mouse model was used and UA was administered orally for 8 weeks. Dexamethasone was used as a positive control. After 8 weeks of administration, the spleen-to-body-weight ratio, renal function, urine albumin excretion, cytokines levels, and the deposition of immune complex were measured. The primary mouse glomerular mesangial cells (GMCs) and SV40-MES-13 were stimulated by lipopolysaccharide (LPS), either alone or in combination with nigericin, to establish an in vitro model. The activation of NLRP3 inflammasome were investigated both in vivo and in vitro using qRT-PCR, immunoblotting, and immunofluorescence. RESULTS: Our results revealed that UA prominently alleviated LN in MRL/lpr lupus-prone mice, leading to a significant reduction in proteinuria production, infiltration of immune cells infiltration, and histopathological damage in the renal tissue. In addition, UA exerted inhibitory effects on the secretion of IL-1ß, IL-18, and caspase-1, pyroptosis, and ASC speck formation in primary mouse GMCs and SV40-MES-13 cells. Furthermore, UA facilitated the degradation of NLRP3 by suppressing SUMO1-mediated SUMOylation of NLRP3. CONCLUSION: UA possess a therapeutical effect on LN in MRL/lpr mice by enhancing the degradation of NLRP3 through inhibition of SUMO1-mediated SUMOylation of NLRP3. Our findings provide a basis for proposing UA as a potential candidate for the treatment of LN.


Subject(s)
Inflammasomes , Lupus Nephritis , Mice, Inbred MRL lpr , NLR Family, Pyrin Domain-Containing 3 Protein , Triterpenes , Ursolic Acid , Animals , Triterpenes/pharmacology , Lupus Nephritis/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mice , Inflammasomes/metabolism , Inflammasomes/drug effects , Disease Models, Animal , Female , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Anti-Inflammatory Agents/pharmacology , Sumoylation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...