Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.361
Filter
1.
Sci Rep ; 14(1): 14972, 2024 06 28.
Article in English | MEDLINE | ID: mdl-38951571

ABSTRACT

Sepsis, marked by organ dysfunction, necessitates reliable biomarkers. Ribonuclease inhibitor 1 (RNH1), a ribonuclease (RNase) inhibitor, emerged as a potential biomarker for acute kidney injury and mortality in thoracoabdominal aortic aneurysm patients. Our study investigates RNH1 dynamics in sepsis, its links to mortality and organ dysfunction, and the interplay with RNase 1 and RNase 5. Furthermore, we explore RNH1 as a therapeutic target in sepsis-related processes like inflammation, non-canonical inflammasome activation, and iron homeostasis. We showed that RNH1 levels are significantly higher in deceased patients compared to sepsis survivors and correlate with creatine kinase, aspartate and alanine transaminase, bilirubin, serum creatinine and RNase 5, but not RNase 1. RNH1 mitigated LPS-induced TNFα and RNase 5 secretion, and relative mRNA expression of ferroptosis-associated genes HMOX1, FTH1 and HAMP in PBMCs. Monocytes were identified as the predominant type of LPS-positive PBMCs. Exogenous RNH1 attenuated LPS-induced CASP5 expression, while increasing IL-1ß secretion in PBMCs and THP-1 macrophages. As RNH1 has contradictory effects on inflammation and non-canonical inflammasome activation, its use as a therapeutic agent is limited. However, RNH1 levels may play a central role in iron homeostasis during sepsis, supporting our clinical observations. Hence, RNH1 shows promise as biomarkers for renal and hepatic dysfunction and hepatocyte injury, and may be useful in predicting the outcome of septic patients.


Subject(s)
Biomarkers , Homeostasis , Inflammation , Iron , Sepsis , Humans , Sepsis/metabolism , Sepsis/drug therapy , Biomarkers/metabolism , Iron/metabolism , Inflammation/metabolism , Male , Female , Middle Aged , Aged , Inflammasomes/metabolism , Lipopolysaccharides , THP-1 Cells , Carrier Proteins
2.
Elife ; 122024 Jul 02.
Article in English | MEDLINE | ID: mdl-38953285

ABSTRACT

We studied lysosomal Ca2+ in inflammasome. Lipopolysaccharide (LPS) + palmitic acid (PA) decreased lysosomal Ca2+ ([Ca2+]Lys) and increased [Ca2+]i through mitochondrial ROS, which was suppressed in Trpm2-KO macrophages. Inflammasome activation and metabolic inflammation in adipose tissue of high-fat diet (HFD)-fed mice were ameliorated by Trpm2 KO. ER→lysosome Ca2+ refilling occurred after lysosomal Ca2+ release whose blockade attenuated LPS + PA-induced inflammasome. Subsequently, store-operated Ca2+entry (SOCE) was activated whose inhibition suppressed inflammasome. SOCE was coupled with K+ efflux whose inhibition reduced ER Ca2+ content ([Ca2+]ER) and impaired [Ca2+]Lys recovery. LPS + PA activated KCa3.1 channel, a Ca2+-activated K+ channel. Inhibitors of KCa3.1 channel or Kcnn4 KO reduced [Ca2+]ER, attenuated increase of [Ca2+]i or inflammasome activation by LPS + PA, and ameliorated HFD-induced inflammasome or metabolic inflammation. Lysosomal Ca2+ release induced delayed JNK and ASC phosphorylation through CAMKII-ASK1. These results suggest a novel role of lysosomal Ca2+ release sustained by ER→lysosome Ca2+ refilling and K+ efflux through KCa3.1 channel in inflammasome activation and metabolic inflammation.


Subject(s)
Calcium , Endoplasmic Reticulum , Inflammasomes , Inflammation , Lysosomes , Mice, Knockout , Potassium , Animals , Inflammasomes/metabolism , Mice , Lysosomes/metabolism , Calcium/metabolism , Potassium/metabolism , Inflammation/metabolism , Endoplasmic Reticulum/metabolism , Lipopolysaccharides , TRPM Cation Channels/metabolism , TRPM Cation Channels/genetics , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Mice, Inbred C57BL , Macrophages/metabolism , Male , Diet, High-Fat
3.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 49(3): 367-376, 2024 Mar 28.
Article in English, Chinese | MEDLINE | ID: mdl-38970510

ABSTRACT

Acute kidney injury (AKI) is a common critical condition in clinical practice, characterized by a rapid decline in renal function within a short period. The pathogenesis of AKI is complex and has not been fully elucidated. In recent years, studies have found that the activation of endoplasmic reticulum stress (ERS) and the Nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome are closely related to the occurrence of AKI. When the kidneys is damaged, the internal environment of the kidney cells is disrupted, leading to the activation of ERS. Excessive ERS can induce apoptosis of renal cells, leading to the occurrence of AKI. Additionally, the NLRP3 inflammasome can mediate the recognition of endogenous and exogenous danger signal molecules by the host, subsequently activating caspase-1, pro-inflammatory cytokines such as IL-1ß and IL-18, inducing inflammatory responses, and promoting apoptosis of renal cells. In animal models of AKI, the upregulation of ERS markers is often accompanied by increased expression levels of NLRP3 inflammasome-related proteins, indicating that ERS can regulate the activation process of the NLRP3 inflammasome. Clarifying the role and mechanism of ERS and NLRP3 inflammasome in AKI is expected to provide new insights for the prevention and treatment of AKI.


Subject(s)
Acute Kidney Injury , Endoplasmic Reticulum Stress , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Acute Kidney Injury/metabolism , Acute Kidney Injury/etiology , Endoplasmic Reticulum Stress/physiology , Inflammasomes/metabolism , Humans , Animals , Apoptosis , Interleukin-18/metabolism , Kidney/metabolism , Interleukin-1beta/metabolism
4.
Cell Commun Signal ; 22(1): 351, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38970061

ABSTRACT

BACKGROUND: Accompanied by activation of the NOD-like receptor protein 3 (NLRP3) inflammasome, aberrant connexin 43 (Cx43) hemichannel-mediated ATP release is situated upstream of inflammasome assembly and inflammation and contributes to multiple secondary complications of diabetes and associated cardiometabolic comorbidities. Evidence suggests there may be a link between Cx43 hemichannel activity and inflammation in the diabetic kidney. The consequences of blocking tubular Cx43 hemichannel-mediated ATP release in priming/activation of the NLRP3 inflammasome in a model of diabetic kidney disease (DKD) was investigated. We examined downstream markers of inflammation and the proinflammatory and chemoattractant role of the tubular secretome on macrophage recruitment and activation. METHODS: Analysis of human transcriptomic data from the Nephroseq repository correlated gene expression to renal function in DKD. Primary human renal proximal tubule epithelial cells (RPTECs) and monocyte-derived macrophages (MDMs) were cultured in high glucose and inflammatory cytokines as a model of DKD to assess Cx43 hemichannel activity, NLRP3 inflammasome activation and epithelial-to-macrophage paracrine-mediated crosstalk. Tonabersat assessed a role for Cx43 hemichannels. RESULTS: Transcriptomic analysis from renal biopsies of patients with DKD showed that increased Cx43 and NLRP3 expression correlated with declining glomerular filtration rate (GFR) and increased proteinuria. In vitro, Tonabersat blocked glucose/cytokine-dependant increases in Cx43 hemichannel-mediated ATP release and reduced expression of inflammatory markers and NLRP3 inflammasome activation in RPTECs. We observed a reciprocal relationship in which NLRP3 activity exacerbated increased Cx43 expression and hemichannel-mediated ATP release, events driven by nuclear factor kappa-B (NFκB)-mediated priming and Cx43 hemichannel opening, changes blocked by Tonabersat. Conditioned media (CM) from RPTECs treated with high glucose/cytokines increased expression of inflammatory markers in MDMs, an effect reduced when macrophages were pre-treated with Tonabersat. Co-culture using conditioned media from Tonabersat-treated RPTECs dampened macrophage inflammatory marker expression and reduced macrophage migration. CONCLUSION: Using a model of DKD, we report for the first time that high glucose and inflammatory cytokines trigger aberrant Cx43 hemichannel activity, events that instigate NLRP3-induced inflammation in RPTECs and epithelial-to-macrophage crosstalk. Recapitulating observations previously reported in diabetic retinopathy, these data suggest that Cx43 hemichannel blockers (i.e., Tonabersat) may dampen multi-system damage observed in secondary complications of diabetes.


Subject(s)
Diabetic Nephropathies , Inflammasomes , Macrophages , NLR Family, Pyrin Domain-Containing 3 Protein , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Humans , Macrophages/metabolism , Macrophages/drug effects , Inflammasomes/metabolism , Connexin 43/metabolism , Connexin 43/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Adenosine Triphosphate/metabolism , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology
5.
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
6.
Cell Mol Life Sci ; 81(1): 295, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38977508

ABSTRACT

Nod-like receptor family pyrin-containing protein 3 (NLRP3) inflammasome plays a pathologic role in metabolic dysfunction-associated steatohepatitis (MASH), but the molecular mechanism regulating the NLRP3 inflammasome activation in hepatocellular lipotoxicity remains largely unknown. Bromodomain-containing protein 4 (BRD4) has emerged as a key epigenetic reader of acetylated lysine residues in enhancer regions that control the transcription of key genes. The aim of this study is to investigate if and how BRD4 regulated the NLRP3 inflammasome activation and pyroptosis in MASH. Using the AML12 and primary mouse hepatocytes stimulated by palmitic acid (PA) as an in vitro model of hepatocellular lipotoxicity, we found that targeting BRD4 by genetic knockdown or a selective BRD4 inhibitor MS417 protected against hepatosteatosis; and this protective effect was attributed to inhibiting the activation of NLRP3 inflammasome and reducing the expression of Caspase-1, gasdermin D (GSDMD), interleukin (IL)-1ß and IL-6. Moreover, BRD4 inhibition limited the voltage-dependent anion channel-1 (VDAC1) expression and oligomerization in PA-treated AML12 hepatocytes, thereby suppressing the NLRP3 inflammasome activation. Additionally, the expression of BRD4 enhanced in MASH livers of humans. Mechanistically, BRD4 was upregulated during hepatocellular lipotoxicity that in turn modulated the active epigenetic mark H3K27ac at the promoter regions of the Vdac and Gsdmd genes, thereby enhancing the expression of VDAC and GSDMD. Altogether, our data provide novel insights into epigenetic mechanisms underlying BRD4 activating the NLRP3 inflammasome and promoting GSDMD-mediated pyroptosis in hepatocellular lipotoxicity. Thus, BRD4 might serve as a novel therapeutic target for the treatment of MASH.


Subject(s)
Hepatocytes , Inflammasomes , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Phosphate-Binding Proteins , Pyroptosis , Transcription Factors , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Hepatocytes/metabolism , Hepatocytes/drug effects , Hepatocytes/pathology , Pyroptosis/drug effects , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/genetics , Inflammasomes/metabolism , Mice , Transcription Factors/metabolism , Transcription Factors/genetics , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Palmitic Acid/pharmacology , Male , Indenes/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Sulfonamides/pharmacology , Fatty Liver/metabolism , Fatty Liver/pathology , Cell Cycle Proteins , Furans , Gasdermins , Bromodomain Containing Proteins , Nuclear Proteins
7.
Int J Mol Sci ; 25(13)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-39000601

ABSTRACT

Chagas disease is caused by the intracellular protozoan parasite Trypanosoma cruzi. This disease affects mainly rural areas in Central and South America, where the insect vector is endemic. However, this disease has become a world health problem since migration has spread it to other continents. It is a complex disease with many reservoirs and vectors and high genetic variability. One of the host proteins involved in the pathogenesis is SLAMF1. This immune receptor acts during the infection of macrophages controlling parasite replication and thus affecting survival in mice but in a parasite strain-dependent manner. Therefore, we studied the role of SLAMF1 by quantitative proteomics in a macrophage in vitro infection and the different responses between Y and VFRA strains of Trypanosoma cruzi. We detected different significant up- or downregulated proteins involved in immune regulation processes, which are SLAMF1 and/or strain-dependent. Furthermore, independently of SLAMF1, this parasite induces different responses in macrophages to counteract the infection and kill the parasite, such as type I and II IFN responses, NLRP3 inflammasome activation, IL-18 production, TLR7 and TLR9 activation specifically with the Y strain, and IL-11 signaling specifically with the VFRA strain. These results have opened new research fields to elucidate the concrete role of SLAMF1 and discover new potential therapeutic approaches for Chagas disease.


Subject(s)
Chagas Disease , Macrophages , Proteomics , Trypanosoma cruzi , Trypanosoma cruzi/metabolism , Animals , Mice , Macrophages/metabolism , Macrophages/parasitology , Macrophages/immunology , Proteomics/methods , Chagas Disease/parasitology , Chagas Disease/metabolism , Chagas Disease/immunology , Antigens, CD/metabolism , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 9/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Interleukin-18/metabolism , Receptors, Cell Surface/metabolism , Inflammasomes/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1 , Membrane Glycoproteins
8.
Zool Res ; 45(4): 857-874, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39004863

ABSTRACT

Emerging evidence indicates that sleep deprivation (SD) can lead to Alzheimer's disease (AD)-related pathological changes and cognitive decline. However, the underlying mechanisms remain obscure. In the present study, we identified the existence of a microbiota-gut-brain axis in cognitive deficits resulting from chronic SD and revealed a potential pathway by which gut microbiota affects cognitive functioning in chronic SD. Our findings demonstrated that chronic SD in mice not only led to cognitive decline but also induced gut microbiota dysbiosis, elevated NLRP3 inflammasome expression, GSK-3ß activation, autophagy dysfunction, and tau hyperphosphorylation in the hippocampus. Colonization with the "SD microbiota" replicated the pathological and behavioral abnormalities observed in chronic sleep-deprived mice. Remarkably, both the deletion of NLRP3 in NLRP3 -/- mice and specific knockdown of NLRP3 in the hippocampus restored autophagic flux, suppressed tau hyperphosphorylation, and ameliorated cognitive deficits induced by chronic SD, while GSK-3ß activity was not regulated by the NLRP3 inflammasome in chronic SD. Notably, deletion of NLRP3 reversed NLRP3 inflammasome activation, autophagy deficits, and tau hyperphosphorylation induced by GSK-3ß activation in primary hippocampal neurons, suggesting that GSK-3ß, as a regulator of NLRP3-mediated autophagy dysfunction, plays a significant role in promoting tau hyperphosphorylation. Thus, gut microbiota dysbiosis was identified as a contributor to chronic SD-induced tau pathology via NLRP3-mediated autophagy dysfunction, ultimately leading to cognitive deficits. Overall, these findings highlight GSK-3ß as a regulator of NLRP3-mediated autophagy dysfunction, playing a critical role in promoting tau hyperphosphorylation.


Subject(s)
Autophagy , Dysbiosis , Gastrointestinal Microbiome , NLR Family, Pyrin Domain-Containing 3 Protein , Sleep Deprivation , tau Proteins , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Gastrointestinal Microbiome/physiology , Sleep Deprivation/metabolism , Sleep Deprivation/physiopathology , Sleep Deprivation/complications , Mice , Autophagy/physiology , tau Proteins/metabolism , tau Proteins/genetics , Male , Hippocampus/metabolism , Mice, Inbred C57BL , Mice, Knockout , Inflammasomes/metabolism
9.
J Am Heart Assoc ; 13(14): e032904, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38979831

ABSTRACT

BACKGROUND: Cardiac aging represents an independent risk factor for aging-associated cardiovascular diseases. Although evidence suggests an association between NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome formation and numerous cardiovascular diseases, its role in cardiac aging remains largely unclear. METHODS AND RESULTS: The longevity of mice with wild-type and NLRP3 knockout (NLRP3-/-) genotypes was assessed, with or without d-galactose treatment. Cardiac function was evaluated using echocardiography, and cardiac histopathology was examined through hematoxylin and eosin and Masson's trichrome staining. Senescence-associated ß-galactosidase (SA-ß-gal) staining was employed to detect cardiac aging. Western blotting was used to assess aging-related proteins (p53, p21) and pyroptosis-related proteins. Additionally, dihydroethidium staining, lactate dehydrogenase release, and interleukin-1ß ELISA assays were performed, along with measurements of total superoxide dismutase and malondialdehyde levels. In vitro, H9c2 cells were exposed to d-galactose for 24 hours in the absence or presence of N-acetyl-l-cysteine (reactive oxygen species inhibitor), BAY-117082 (nuclear factor κ-light-chain enhancer of activated B cells inhibitor), MCC950 (NLRP3 inhibitor), and VX-765 (Caspase-1 inhibitor). Immunofluorescence staining was employed to detect p53, gasdermin D, and apoptosis-associated speck-like protein proteins. Intracellular reactive oxygen species levels were assessed using fluorescence microscopy and flow cytometry. Senescence-associated ß-galactosidase staining and Western blotting were also employed in vitro for the same purpose. The results showed that NLRP3 upregulation was implicated in aging and cardiovascular diseases. Inhibition of NLRP3 extended life span, mitigated the aging phenotype, improved cardiac function and blood pressure, ameliorated lipid metabolism abnormalities, inhibited pyroptosis in cardiomyocytes, and ultimately alleviated cardiac aging. In vitro, the inhibition of reactive oxygen species, nuclear factor κ-light-chain enhancer of activated B cells, NLRP3, or caspase-1 attenuated NLRP3 inflammasome-mediated pyroptosis. CONCLUSIONS: The reactive oxygen species/nuclear factor κ-light-chain enhancer of activated B cells/NLRP3 signaling pathway loop contributes to d-galactose-treated cardiomyocyte senescence and cardiac aging.


Subject(s)
Galactose , Inflammasomes , Mice, Knockout , Myocytes, Cardiac , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Galactose/toxicity , Galactose/metabolism , Pyroptosis/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Inflammasomes/metabolism , Mice , Aging/metabolism , Mice, Inbred C57BL , Signal Transduction , Cellular Senescence/drug effects , Male , Reactive Oxygen Species/metabolism , NF-kappa B/metabolism , Cell Line , Disease Models, Animal , Rats
10.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000248

ABSTRACT

Antiretroviral treatment (ART) has converted HIV from a lethal disease to a chronic condition, yet co-morbidities persist. Incomplete immune recovery and chronic immune activation, especially in the gut mucosa, contribute to these complications. Inflammasomes, multi-protein complexes activated by innate immune receptors, appear to play a role in these inflammatory responses. In particular, preliminary data indicate the involvement of IFI16 and NLRP3 inflammasomes in chronic HIV infection. This study explores inflammasome function in monocytes from people with HIV (PWH); 22 ART-treated with suppressed viremia and 17 untreated PWH were compared to 33 HIV-negative donors. Monocytes were primed with LPS and inflammasomes activated with ATP in vitro. IFI16 and NLRP3 mRNA expression were examined in a subset of donors. IFI16 and NLRP3 expression in unstimulated monocytes correlated negatively with CD4 T cell counts in untreated PWH. For IFI16, there was also a positive correlation with viral load. Monocytes from untreated PWH exhibit increased release of IL-1α, IL-1ß, and TNF compared to treated PWH and HIV-negative donors. However, circulating monocytes in PWH are not pre-primed for inflammasome activation in vivo. The findings suggest a link between IFI16, NLRP3, and HIV progression, emphasizing their potential role in comorbidities such as cardiovascular disease. The study provides insights into inflammasome regulation in HIV pathogenesis and its implications for therapeutic interventions.


Subject(s)
HIV Infections , Inflammasomes , Interleukin-1alpha , Interleukin-1beta , Monocytes , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Monocytes/metabolism , Monocytes/immunology , HIV Infections/immunology , HIV Infections/virology , HIV Infections/metabolism , Interleukin-1beta/metabolism , Inflammasomes/metabolism , Male , Female , Adult , Middle Aged , Interleukin-1alpha/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Phosphoproteins/metabolism , Chronic Disease , Viral Load
12.
BMC Cardiovasc Disord ; 24(1): 354, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38992615

ABSTRACT

BACKGROUND: Hyperlipidemia damages vascular wall and serves as a foundation for diseases such as atherosclerosis, hypertension and stiffness. The NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome is implicated in vascular dysfunction associated with hyperlipidemia-induced vascular injury. Sodium tanshinone IIA sulfonate (STS), a well-established cardiovascular protective drug with recognized anti-inflammatory, antioxidant, and vasodilatory properties, is yet to be thoroughly investigated for its impact on vascular relaxant imbalance induced by hyperlipidemia. METHODS: In this study, we treated ApoE-knockout (ApoE-/-) mouse with STS and assessed the activation of the NLRP3 inflammasome, expression of MMP2/9, integrity of elastic fibers, and vascular constriction and relaxation. RESULTS: Our findings reveal that STS intervention effectively preserves elastic fibers, significantly restores aortic relaxation function in ApoE-/- mice, and reduces their excessive constriction. Furthermore, STS inhibits the phosphorylation of spleen tyrosine kinase (SYK), suppresses NLRP3 inflammasome activation, and reduces MMP2/9 expression. CONCLUSIONS: These results demonstrate that STS protects vascular relaxation against hyperlipidemia-induced damage through modulation of the SYK-NLRP3 inflammasome-MMP2/9 pathway. This research provides novel insights into the mechanisms underlying vascular relaxation impairment in a hyperlipidemic environment and uncovers a unique mechanism by which STS preserves vascular relaxation, offering valuable foundational research evidence for its clinical application in promoting vascular health.


Subject(s)
Disease Models, Animal , Inflammasomes , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , Mice, Inbred C57BL , Mice, Knockout, ApoE , NLR Family, Pyrin Domain-Containing 3 Protein , Phenanthrenes , Signal Transduction , Syk Kinase , Vasodilation , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Syk Kinase/metabolism , Matrix Metalloproteinase 2/metabolism , Phenanthrenes/pharmacology , Male , Matrix Metalloproteinase 9/metabolism , Vasodilation/drug effects , Hyperlipidemias/drug therapy , Hyperlipidemias/physiopathology , Vasodilator Agents/pharmacology , Phosphorylation , Mice , Aorta/drug effects , Aorta/physiopathology , Aorta/metabolism , Aorta/enzymology , Apolipoproteins E
13.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(6): 1024-1032, 2024 Jun 20.
Article in Chinese | MEDLINE | ID: mdl-38977331

ABSTRACT

OBJECTIVE: To investigate the mechanism of 2, 6-dimethoxy-1, 4-benzoquinone (DMQ), an active ingredients in fermented wheat germ extract, for inhibiting NLRP3 inflammasome activation and alleviating septic shock in mice. METHODS: Cultured murine bone marrow-derived macrophages (BMDM) stimulated with lipopolysaccharide (LPS) were treated with DMQ, followed by treatment with Nigericin, ATP, and MSU for activating the canonical NLRP3 inflammasome; the noncanonical NLRP3 inflammasome was activated by intracellular transfection of LPS, and AIM2 inflammasome was activated using Poly A: T.In human monocytic THP-1 cells, the effect of Nigericin on inflammasome activation products was examined using Western blotting and ELISA.Co-immunoprecipitation was performed to explore the mechanism of DMQ-induced blocking of NLRP3 inflammasome activation.In a male C57BL/6J mouse model of LPS-induced septic shock treated with 20 and 40 mg/kg DMQ, the levels of IL-1ß and TNF-α in the serum and peritoneal lavage fluid were determined using ELISA, and the survival time of the mice within 36 h was observed. RESULTS: Treatment with DMQ effectively inhibited LPS-induced activation of canonical NLRP3 inflammasome in mouse BMDM and human THP-1 cells and also inhibited non-canonical NLRP3 inflammasome activation in mouse BMDM, but produced no significant effect on AIM2 inflammasome activation.DMQ significantly blocked the binding between ASC and NLRP3.In the mouse models of septic shock, DMQ treatment significantly reduced the levels of IL-1ß in the serum and peritoneal fluid and obviously prolonged survival time of the mice. CONCLUSION: DMQ can effectively block ASC-NLRP3 interaction to inhibit NLRP3 inflammasome activation and alleviate LPSinduced septic shock in mice.


Subject(s)
Benzoquinones , Inflammasomes , Interleukin-1beta , Lipopolysaccharides , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Shock, Septic , Animals , Shock, Septic/drug therapy , Shock, Septic/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mice , Inflammasomes/metabolism , Male , Humans , Benzoquinones/pharmacology , Benzoquinones/therapeutic use , Interleukin-1beta/metabolism , Macrophages/metabolism , Macrophages/drug effects , Tumor Necrosis Factor-alpha/metabolism , THP-1 Cells , Disease Models, Animal
14.
BMC Cardiovasc Disord ; 24(1): 351, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987672

ABSTRACT

Diabetic cardiomyopathy (DCM) is a chronic disease caused by diabetes mellitus, which is recognized as a worldwide challenging disease. This study aimed to investigate the role and the potential mechanism of knocking down the NACHT-, LRR- and PYD domains-containing protein 3 (NLRP3), an inflammasome associated with onset and progression of various diseases, on high glucose or diabetes -induced cardiac cells pyroptosis and ferroptosis, two regulated non-necrosis cell death modalities discovered recent years. In the present study, both in vivo and in vitro studies were conducted simultaneously. Diabetic rats were induced by 55 mg/kg intraperitoneal injection of streptozotocin (STZ). Following the intraperitoneal injection of MCC950 (10 mg/kg), On the other hand, the DCM model in H9C2 cardiac cells was simulated with 35 mmol/L glucose and a short hairpin RNA vector of NLRP3 were transfected to cells. The results showed that in vivo study, myocardial fibers were loosely arranged and showed inflammatory cell infiltration, mitochondrial cristae were broken and the GSDMD-NT expression was found notably increased in the DM group, while the protein expressions of xCT and GPX4 was significantly decreased, both of which were reversed by MCC950. High glucose reduced the cell viability and ATP level in vitro, accompanied by an increase in LDH release. All of the above indicators were reversed after NLRP3 knockdown compared with the HG treated alone. Moreover, the protein expressions of pyroptosis- and ferroptosis-related fators were significantly decreased or increased, consistent with the results shown by immunofluorescence. Furthermore, the protective effects of NLRP3 knockdown against HG were reversed following the mtROS agonist rotenone (ROT) treatment. In conclusion, inhibition of NLRP3 suppressed DM-induced myocardial injury. Promotion of mitochondrial ROS abolished the protective effect of knockdown NLRP3, and induced the happening of pyroptosis and ferroptosis. These findings may present a novel therapeutic underlying mechanism for clinical diabetes-induced myocardial injury treatment.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies , Ferroptosis , Gene Knockdown Techniques , Myocytes, Cardiac , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Animals , Ferroptosis/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Male , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Cell Line , Rats, Sprague-Dawley , Rats , Signal Transduction , Reactive Oxygen Species/metabolism , Inflammasomes/metabolism , Sulfonamides/pharmacology , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/genetics , Gasdermins
15.
Proc Natl Acad Sci U S A ; 121(29): e2400883121, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38980908

ABSTRACT

Gasdermin D (GSDMD)-mediated pyroptotic cell death drives inflammatory cytokine release and downstream immune responses upon inflammasome activation, which play important roles in host defense and inflammatory disorders. Upon activation by proteases, the GSDMD N-terminal domain (NTD) undergoes oligomerization and membrane translocation in the presence of lipids to assemble pores. Despite intensive studies, the molecular events underlying the transition of GSDMD from an autoinhibited soluble form to an oligomeric pore form inserted into the membrane remain incompletely understood. Previous work characterized S-palmitoylation for gasdermins from bacteria, fungi, invertebrates, as well as mammalian gasdermin E (GSDME). Here, we report that a conserved residue Cys191 in human GSDMD was S-palmitoylated, which promoted GSDMD-mediated pyroptosis and cytokine release. Mutation of Cys191 or treatment with palmitoyltransferase inhibitors cyano-myracrylamide (CMA) or 2-bromopalmitate (2BP) suppressed GSDMD palmitoylation, its localization to the membrane and dampened pyroptosis or IL-1ß secretion. Furthermore, Gsdmd-dependent inflammatory responses were alleviated by inhibition of palmitoylation in vivo. By contrast, coexpression of GSDMD with palmitoyltransferases enhanced pyroptotic cell death, while introduction of exogenous palmitoylation sequences fully restored pyroptotic activities to the C191A mutant, suggesting that palmitoylation-mediated membrane localization may be distinct from other molecular events such as GSDMD conformational change during pore assembly. Collectively, our study suggests that S-palmitoylation may be a shared regulatory mechanism for GSDMD and other gasdermins, which points to potential avenues for therapeutically targeting S-palmitoylation of gasdermins in inflammatory disorders.


Subject(s)
Cysteine , Intracellular Signaling Peptides and Proteins , Lipoylation , Phosphate-Binding Proteins , Pyroptosis , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/genetics , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Cysteine/metabolism , Animals , Mice , Cytokines/metabolism , HEK293 Cells , Inflammasomes/metabolism , Gasdermins
16.
Sci Signal ; 17(845): eadd8913, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39012939

ABSTRACT

Hypoxia and low glucose abundance often occur simultaneously at sites of inflammation. In monocytes and macrophages, glucose-oxygen deprivation stimulates the assembly of the NLRP3 inflammasome to generate the proinflammatory cytokine IL-1ß. We found that concomitant glucose deprivation and hypoxia activated the NLRP3 inflammasome by constraining the function of HMG-CoA reductase (HMGCR), the rate-limiting enzyme of the mevalonate kinase pathway. HMGCR is involved in the synthesis of geranylgeranyl pyrophosphate (GGPP), which is required for the prenylation and lipid membrane integration of proteins. Under glucose-oxygen deprivation, GGPP synthesis was decreased, leading to reduced prenylation of the small GTPase Rac1, increased binding of nonprenylated Rac1 to the scaffolding protein IQGAP1, and enhanced activation of the NLRP3 inflammasome. In response to restricted oxygen and glucose supply, patient monocytes with a compromised mevalonate pathway due to mevalonate kinase deficiency or Muckle-Wells syndrome released more IL-1ß than did control monocytes. Thus, reduced GGPP synthesis due to inhibition of HMGCR under glucose-oxygen deprivation results in proinflammatory innate responses, which are normally kept in check by the prenylation of Rac1. We suggest that this mechanism is also active in inflammatory autoimmune conditions.


Subject(s)
Glucose , Hydroxymethylglutaryl CoA Reductases , Inflammasomes , Monocytes , NLR Family, Pyrin Domain-Containing 3 Protein , rac1 GTP-Binding Protein , Humans , rac1 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/genetics , Monocytes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Hydroxymethylglutaryl CoA Reductases/metabolism , Hydroxymethylglutaryl CoA Reductases/genetics , Inflammasomes/metabolism , Glucose/metabolism , Polyisoprenyl Phosphates/metabolism , Interleukin-1beta/metabolism , Oxygen/metabolism , Protein Prenylation , Mevalonate Kinase Deficiency/metabolism , Mevalonate Kinase Deficiency/genetics , Mevalonic Acid/metabolism
17.
Theranostics ; 14(10): 3945-3962, 2024.
Article in English | MEDLINE | ID: mdl-38994035

ABSTRACT

Rationale: NLRP3 inflammasome is critical in the development and progression of many metabolic diseases driven by chronic inflammation, but its effect on the pathology of postmenopausal osteoporosis (PMOP) remains poorly understood. Methods: We here firstly examined the levels of NLRP3 inflammasome in PMOP patients by ELISA. Then we investigated the possible mechanisms underlying the effect of NLRP3 inflammasome on PMOP by RNA sequencing of osteoblasts treated with NLRP3 siRNA and qPCR. Lastly, we accessed the effect of decreased NLRP3 levels on ovariectomized (OVX) rats. To specifically deliver NLRP3 siRNA to osteoblasts, we constructed NLRP3 siRNA wrapping osteoblast-specific aptamer (CH6)-functionalized lipid nanoparticles (termed as CH6-LNPs-siNLRP3). Results: We found that the levels of NLRP3 inflammasome were significantly increased in patients with PMOP, and were negatively correlated with estradiol levels. NLRP3 knock-down influenced signal pathways including immune system process, interferon signal pathway. Notably, of the top ten up-regulated genes in NLRP3-reduced osteoblasts, nine genes (except Mx2) were enriched in immune system process, and five genes were related to interferon signal pathway. The in vitro results showed that CH6-LNPs-siNLRP3 was relatively uniform with a dimeter of 96.64 ± 16.83 nm and zeta potential of 38.37 ± 1.86 mV. CH6-LNPs-siNLRP3 did not show obvious cytotoxicity and selectively delivered siRNA to bone tissue. Moreover, CH6-LNPs-siNLRP3 stimulated osteoblast differentiation by activating ALP and enhancing osteoblast matrix mineralization. When administrated to OVX rats, CH6-LNPs-siNLRP3 promoted bone formation and bone mass, improved bone microarchitecture and mechanical properties by decreasing the levels of NLRP3, IL-1ß and IL-18 and increasing the levels of OCN and Runx2. Conclusion: NLRP3 inflammasome may be a new biomarker for PMOP diagnosis and plays a key role in the pathology of PMOP. CH6-LNPs-siNLRP3 has potential application for the treatment of PMOP.


Subject(s)
Inflammasomes , Liposomes , NLR Family, Pyrin Domain-Containing 3 Protein , Nanoparticles , Osteoblasts , Osteoporosis, Postmenopausal , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Female , Humans , Rats , Inflammasomes/metabolism , Nanoparticles/chemistry , Osteoporosis, Postmenopausal/metabolism , Down-Regulation/drug effects , Rats, Sprague-Dawley , RNA, Small Interfering/administration & dosage , Aptamers, Nucleotide/pharmacology , Aptamers, Nucleotide/administration & dosage , Disease Models, Animal , Middle Aged , Ovariectomy
18.
Exp Dermatol ; 33(7): e15135, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39021278

ABSTRACT

Autoimmune skin disease is a kind of heterogeneous disease with complicated pathogenesis. Many factors such as genetic, infectious, environmental and even psychological factors may interact together to trigger a synergistic effect for the development of abnormal innate and adaptive immune responses. Although the exact mechanisms remain unclear, recent evidence suggests that pyroptosis plays a pivotal role in the development of autoimmune skin disease. The feature of pyroptosis is the first formation of pores in cellular membranes, then cell rupture and the release of intracellular substances and pro-inflammatory cytokines, such as interleukin-1 beta (IL-1ß) and IL-18. This hyperactive inflammatory programmed cell death damages the homeostasis of the immune system and advances autoimmunity. This review briefly summarises the molecular regulatory mechanisms of pyrin domain-containing protein 3 (NLRP3) inflammasome and gasdermin family, as well as the molecular mechanisms of pyroptosis, highlights the latest progress of pyroptosis in autoimmune skin disease, including systemic lupus erythematosus, psoriasis, atopic dermatitis and systemic scleroderma and attempts to identify its potential advantages as a therapeutic target or prognostic biomarker for these diseases.


Subject(s)
Autoimmune Diseases , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Humans , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Skin Diseases/immunology , Animals , Phosphate-Binding Proteins/metabolism , Interleukin-1beta/metabolism , Scleroderma, Systemic/immunology , Lupus Erythematosus, Systemic/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Psoriasis/immunology , Psoriasis/metabolism , Autoimmunity , Interleukin-18/metabolism , Dermatitis, Atopic/immunology
19.
Am J Reprod Immunol ; 92(1): e13893, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38958245

ABSTRACT

PROBLEM: Vulvovaginal candidiasis (VVC) is a common mucosal fungal infection, and Candida albicans is the main causative agent. The NLRP3 inflammasome plays an important role in VVC, but the underlying mechanism is unknown. METHOD OF STUDY: Vaginal epithelial cells were divided into three groups: control, C. albicans strain SC5314 (wild-type, WT), and WT+ Matt Cooper Compound 950 (MCC950, a specific NLRP3 inhibitor). After human vaginal epithelial cells were pretreated with 1 µmol/L MCC950 for 2 h, C. albicans (MOI = 1) was cocultured with the human vaginal epithelial cells for 12 h. The cell supernatants were collected, LDH was detected, and the IL-1ß and IL-18 levels were determined by ELISA. The expression of the pyroptosis-related proteins NLRP3, Caspase-1 p20 and GSDMD was measured by Western blotting analysis. The protein expression of the pyroptosis-related N-terminus of GSDMD (GSDMD-N) was detected by immunofluorescence. RESULTS: In this study, we showed that the WT C. albicans strain induced pyroptosis in vaginal epithelial cells, as indicated by the LDH and proinflammatory cytokine levels and the upregulated levels of the pyroptosis-related proteins NLRP3, Caspase-1 p20, and GSDMD-N. MCC950 reversed the changes in the expression of these proteins and proinflammatory cytokines in vaginal epithelial cells. CONCLUSION: C. albicans activated the NLRP3 inflammasome to induce vaginal epithelial cell pyroptosis. MCC950 inhibited the NLRP3 inflammasome, reduced vaginal epithelial cell pyroptosis, and decreased the release of inflammatory cytokines.


Subject(s)
Candida albicans , Candidiasis, Vulvovaginal , Epithelial Cells , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Vagina , Female , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Candidiasis, Vulvovaginal/immunology , Candidiasis, Vulvovaginal/microbiology , Candidiasis, Vulvovaginal/metabolism , Epithelial Cells/immunology , Epithelial Cells/metabolism , Inflammasomes/metabolism , Inflammasomes/immunology , Candida albicans/immunology , Vagina/microbiology , Vagina/immunology , Vagina/pathology , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Indenes , Furans/pharmacology , Caspase 1/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Phosphate-Binding Proteins/metabolism , Cells, Cultured , Sulfonamides
SELECTION OF CITATIONS
SEARCH DETAIL
...