Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
J Am Heart Assoc ; 13(9): e034731, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38700011

ABSTRACT

BACKGROUND: Cardiac damage induced by ischemic stroke, such as arrhythmia, cardiac dysfunction, and even cardiac arrest, is referred to as cerebral-cardiac syndrome (CCS). Cardiac macrophages are reported to be closely associated with stroke-induced cardiac damage. However, the role of macrophage subsets in CCS is still unclear due to their heterogeneity. Sympathetic nerves play a significant role in regulating macrophages in cardiovascular disease. However, the role of macrophage subsets and sympathetic nerves in CCS is still unclear. METHODS AND RESULTS: In this study, a middle cerebral artery occlusion mouse model was used to simulate ischemic stroke. ECG and echocardiography were used to assess cardiac function. We used Cx3cr1GFPCcr2RFP mice and NLRP3-deficient mice in combination with Smart-seq2 RNA sequencing to confirm the role of macrophage subsets in CCS. We demonstrated that ischemic stroke-induced cardiac damage is characterized by severe cardiac dysfunction and robust infiltration of monocyte-derived macrophages into the heart. Subsequently, we identified that cardiac monocyte-derived macrophages displayed a proinflammatory profile. We also observed that cardiac dysfunction was rescued in ischemic stroke mice by blocking macrophage infiltration using a CCR2 antagonist and NLRP3-deficient mice. In addition, a cardiac sympathetic nerve retrograde tracer and a sympathectomy method were used to explore the relationship between sympathetic nerves and cardiac macrophages. We found that cardiac sympathetic nerves are significantly activated after ischemic stroke, which contributes to the infiltration of monocyte-derived macrophages and subsequent cardiac dysfunction. CONCLUSIONS: Our findings suggest a potential pathogenesis of CCS involving the cardiac sympathetic nerve-monocyte-derived macrophage axis.


Subject(s)
Disease Models, Animal , Ischemic Stroke , Macrophages , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Ischemic Stroke/physiopathology , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Male , Mice, Knockout , Mice , Infarction, Middle Cerebral Artery/physiopathology , Infarction, Middle Cerebral Artery/pathology , Sympathetic Nervous System/physiopathology , Myocardium/pathology , Myocardium/metabolism , Heart Diseases/etiology , Heart Diseases/physiopathology , Heart Diseases/pathology , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/deficiency
2.
Immunology ; 165(2): 195-205, 2022 02.
Article in English | MEDLINE | ID: mdl-34773253

ABSTRACT

Although sepsis in burn patients is a major contributor to mortality, treatments are not always effective and underlying mechanisms have yet to be completely elucidated. NLRP3 inflammasome orchestrates burn-induced, inflammatory-driven pathophysiologic processes. Here, we determined the mechanism of NLRP3 inflammasome activation on bacterial clearance and mortality in burn sepsis. We obtained tissue and blood from 30 wild-type and 30 Nlrp3-/- mice. Mice were subjected to a two-hit model of 25-30% TBSA scald burn followed by Pseudomonas aeruginosa wound infection 72 hours after injury. We also obtained tissue from 34 adult burn patients (≥18 years of age) with early (0-11 days post-burn) and later (≥12 days post-burn) surgical time-points and ten healthy controls. Murine studies indicated that Nlrp3-/- had 30% improved survival and bacterial clearance at the site of injury and is systemically relative to burn sepsis wild type. Greater macrophage and neutrophil infiltration occurred acutely after infection (12 hours) to the site of injury and adipose tissue. This was followed by increased macrophage and neutrophil infiltration to lymphoid organs and liver beyond the acute phase (24 and 72 hours). Interestingly, Nlrp3 ablation increased acute systemic inflammation (IL-6, TNF-α, IL-1ß). Septic burn patients had persistently increased adipose NLRP3 by-product expression beyond the acute phase that was more pronounced in late-onset sepsis. Our findings suggest that Nlrp3 genetic ablation enhanced acute tissue-specific inflammatory responsiveness. Likely, this occurs by paradoxically increasing acute immune infiltration and inflammation with a non-persistent response. Clinically, persistent NLRP3-mediated inflammation occurs in septic versus normal burn patients and potentially detrimentally impacts patient outcomes.


Subject(s)
Burns/complications , Disease Susceptibility , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Sepsis/etiology , Sepsis/metabolism , Animals , Cytokines/metabolism , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Organ Specificity , Prognosis , Sepsis/mortality , Sepsis/therapy
3.
Sci Rep ; 11(1): 24432, 2021 12 24.
Article in English | MEDLINE | ID: mdl-34952919

ABSTRACT

Despite the initial success of some drugs and vaccines targeting COVID-19, understanding the mechanism underlying SARS-CoV-2 disease pathogenesis remains crucial for the development of further approaches to treatment. Some patients with severe Covid-19 experience a cytokine storm and display evidence of inflammasome activation leading to increased levels of IL-1ß and IL-18; however, other reports have suggested reduced inflammatory responses to Sars-Cov-2. In this study we have examined the effects of the Sars-Cov-2 envelope (E) protein, a virulence factor in coronaviruses, on inflammasome activation and pulmonary inflammation. In cultured macrophages the E protein suppressed inflammasome priming and NLRP3 inflammasome activation. Similarly, in mice transfected with E protein and treated with poly(I:C) to simulate the effects of viral RNA, the E protein, in an NLRP3-dependent fashion, reduced expression of pro-IL-1ß, levels of IL-1ß and IL-18 in broncho-alveolar lavage fluid, and macrophage infiltration in the lung. To simulate the effects of more advanced infection, macrophages were treated with both LPS and poly(I:C). In this setting the E protein increased NLRP3 inflammasome activation in both murine and human macrophages. Thus, the Sars-Cov-2 E protein may initially suppress the host NLRP3 inflammasome response to viral RNA while potentially increasing NLRP3 inflammasome responses in the later stages of infection. Targeting the Sars-Cov-2 E protein especially in the early stages of infection may represent a novel approach to Covid-19 therapy.


Subject(s)
Coronavirus Envelope Proteins/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , SARS-CoV-2/metabolism , Animals , Bronchoalveolar Lavage Fluid/chemistry , COVID-19/pathology , COVID-19/virology , Coronavirus Envelope Proteins/genetics , Down-Regulation/drug effects , Endoplasmic Reticulum Stress , Humans , Inflammasomes/drug effects , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Janus Kinases/genetics , Janus Kinases/metabolism , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Poly I-C/pharmacology , RNA, Viral/metabolism , SARS-CoV-2/drug effects , SARS-CoV-2/isolation & purification
4.
Int J Mol Sci ; 22(22)2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34830316

ABSTRACT

Dental calculus (DC) is a common deposit in periodontitis patients. We have previously shown that DC contains both microbial components and calcium phosphate crystals that induce an osteoclastogenic cytokine IL-1ß via the NLRP3 inflammasome in macrophages. In this study, we examined the effects of cytokines produced by mouse macrophages stimulated with DC on osteoclastogenesis. The culture supernatants from wild-type (WT) mouse macrophages stimulated with DC accelerated osteoclastogenesis in RANKL-primed mouse bone marrow macrophages (BMMs), but inhibited osteoclastogenesis in RANKL-primed RAW-D cells. WT, but not NLRP3-deficient, mouse macrophages stimulated with DC produced IL-1ß and IL-18 in a dose-dependent manner, indicating the NLRP3 inflammasome-dependent production of IL-1ß and IL-18. Both WT and NLRP3-deficient mouse macrophages stimulated with DC produced IL-10, indicating the NLRP3 inflammasome-independent production of IL-10. Recombinant IL-1ß accelerated osteoclastogenesis in both RANKL-primed BMMs and RAW-D cells, whereas recombinant IL-18 and IL-10 inhibited osteoclastogenesis. These results indicate that DC induces osteoclastogenic IL-1ß in an NLRP3 inflammasome-dependent manner and anti-osteogenic IL-18 and IL-10 dependently and independently of the NLRP3 inflammasome, respectively. DC may promote alveolar bone resorption via IL-1ß induction in periodontitis patients, but suppress resorption via IL-18 and IL-10 induction in some circumstances.


Subject(s)
Dental Calculus/genetics , Interleukin-10/genetics , Interleukin-18/genetics , Interleukin-1beta/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Osteogenesis/genetics , Alveolar Bone Loss/genetics , Alveolar Bone Loss/immunology , Alveolar Bone Loss/pathology , Animals , Cell Line , Culture Media, Conditioned/pharmacology , Dental Calculus/immunology , Dental Calculus/pathology , Disease Models, Animal , Gene Expression Regulation , Humans , Inflammasomes/drug effects , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-10/immunology , Interleukin-10/pharmacology , Interleukin-18/immunology , Interleukin-18/pharmacology , Interleukin-1beta/immunology , Interleukin-1beta/pharmacology , Macrophage Activation , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Osteoclasts/immunology , Osteoclasts/pathology , Osteogenesis/immunology , Periodontitis/genetics , Periodontitis/immunology , Periodontitis/pathology , Primary Cell Culture , RANK Ligand/genetics , RANK Ligand/immunology , Signal Transduction
5.
Cell Death Dis ; 12(10): 871, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34561424

ABSTRACT

Atopic dermatitis (AD) is a common chronic pruritic inflammatory skin disorder characterized by recurrent eczematous lesions. Interleukin (IL)-33, a cytokine of the IL-1 family, was found to play an important role in the pathogenesis of AD. As a key component of the inflammasome, NLRP3 has been mostly described in myeloid cells that to mediate inflammasome activation conducted proinflammatory cytokine production of the IL-1 family. However, the role of NLRP3 inflammasome in the pathogenesis of AD, as well as IL-33 processing are highly controversial. Whether NLRP3 can mediate IL-33 expression and secretion independently of the inflammasome in the epithelium of AD has remained unclear. In this article, we found the mRNA expression of Il33 and Nlrp3 were notably increased in the lesional skin of AD patients compared to healthy controls. We then found a significant positive correlation between the expression of Nlrp3 and Il33 in the epithelium of MC903-mediated AD mice model, but no changes were observed for Il36α, Il36γ, Il1ß, or Il18 mRNA expression, as well as IL-1ß or IL-18 production. Overexpression of NLRP3 in human immortalized epithelial cells increased IL-33 expression, whereas siRNA targeting NLRP3 abolished IL-33 expression. In addition, inhibition of NLRP3 inflammasome activation or caspase-1 activity with MCC950 or VX-765 showed no effect on the expression and secretion of IL-33 in AD mice. Unlike myeloid cells, NLRP3 predominantly located in the nucleus of epithelial cells, which could directly bind to Il33 specific-promoters and transactivate it through an interaction with transcription factor IRF4. Furthermore, NLRP3 deficient mice exhibited a significant alleviated epidermis inflammation and decreased mRNA expression and secretion of IL-33 in MC903-mediated AD mice without interfering with TSLP and IL-1ß production. Our results demonstrate a novel ability of NLRP3 to function as a crucial transcription factor of IL-33 in epithelium independently of inflammasome that to mediate the pathological process of AD.


Subject(s)
Dermatitis, Atopic/metabolism , Epithelial Cells/metabolism , Inflammasomes/metabolism , Interleukin-33/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Transcription Factors/metabolism , Animals , Calcitriol/analogs & derivatives , Cell Nucleus/metabolism , Dermatitis, Atopic/genetics , Dermatitis, Atopic/pathology , Disease Models, Animal , Gene Expression Regulation , HaCaT Cells , Humans , Interferon Regulatory Factors/metabolism , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Int J Mol Sci ; 22(17)2021 Aug 27.
Article in English | MEDLINE | ID: mdl-34502191

ABSTRACT

Human hepatocellular carcinoma (HCC) is the most common and even worse at prognosis. The patients with HCC which accompanied by other diseases, such as cirrhosis, can be limited in various treatments, such as chemotherapy, not HCC patients without other diseases. NLRP3 inflammasome plays an important role in the innate immune response, but emerging evidence has indicated that the NLRP3 inflammasome is implicated in all stages of cancer development. Various cells express NLRP3 protein through the autocrine or paracrine signaling in their environment, but NK cells do not. The expanding evidence shows that patients who suffer from liver cancers have a low frequency of natural killer (NK) cells, and the function of these cells is also impaired. Thus, we examined how the expression of NLRP3 in HCC cells affects cancer surveillance by NK cells in a state of a co-culture of both cells. When the expression of NLRP3 in HCC cells was ablated, MICA/B on the surface of HCC cells was upregulated through the lowered expression of matrix metalloproteinase. The expression of MICA on the surface of HCC cells interacted with the NKG2D receptor on NK-92 cells, which led to NK cytotoxicity. Furthermore, in a xenograft mice model, NLRP3 KO HCC cells delayed tumor development and metastasis as well as increased the sensitivity to NK cell cytotoxicity. Taken together, NLRP3 KO in HCC could enhance NK immunosurveillance through an interaction of NKG2D-MICA.


Subject(s)
Carcinoma, Hepatocellular/immunology , Cytotoxicity, Immunologic/immunology , Histocompatibility Antigens Class I/metabolism , Killer Cells, Natural/immunology , Monitoring, Immunologic/methods , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , CRISPR-Cas Systems , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class I/genetics , Humans , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred NOD , Mice, SCID , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
J Neuroinflammation ; 18(1): 153, 2021 Jul 06.
Article in English | MEDLINE | ID: mdl-34229722

ABSTRACT

BACKGROUND: Neuroinflammation is a major driver of age-related brain degeneration and concomitant functional impairment. In patients with Alzheimer's disease, the most common form of age-related dementia, factors that enhance neuroinflammation may exacerbate disease progression, in part by impairing the glymphatic system responsible for clearance of pathogenic beta-amyloid. Inflammatory bowel diseases (IBDs) induce neuroinflammation and exacerbate cognitive impairment in the elderly. The NACHT-LRR and pyrin (PYD) domain-containing protein 3 (NLRP3) inflammasome has been implicated in neuroinflammation. Therefore, we examined if the NLRP3 inflammasome contributes to glymphatic dysfunction and cognitive impairment in an aging mouse model of IBD. METHODS: Sixteen-month-old C57BL/6J and NLRP3 knockout (KO) mice received 1% wt/vol dextran sodium sulfate (DSS) in drinking water to model IBD. Colitis induction was confirmed by histopathology. Exploratory behavior was examined in the open field, associative memory by the novel-object recognition and Morris water maze tests, glymphatic clearance by in vivo two-photon imaging, and neuroinflammation by immunofluorescence and western blotting detection of inflammatory markers. RESULTS: Administration of DSS induced colitis, impaired spatial and recognition memory, activated microglia, and increased A1-like astrocyte numbers. In addition, DSS treatment impaired glymphatic clearance, aggravated amyloid plaque accumulation, and induced neuronal loss in the cortex and hippocampus. These neurodegenerative responses were associated with increased NLRP3 inflammasome expression and accumulation of gut-derived T lymphocytes along meningeal lymphatic vessels. Conversely, NLRP3 depletion protected against cognitive dysfunction, neuroinflammation, and neurological damage induced by DSS. CONCLUSIONS: Colitis can exacerbate age-related neuropathology, while suppression of NLRP3 inflammasome activity may protect against these deleterious effects of colitis.


Subject(s)
Brain/metabolism , Cognitive Dysfunction/metabolism , Colitis/metabolism , Inflammation Mediators/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , Age Factors , Animals , Brain/pathology , Chronic Disease , Cognitive Dysfunction/pathology , Colitis/pathology , Female , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency
8.
Front Immunol ; 12: 683803, 2021.
Article in English | MEDLINE | ID: mdl-34122445

ABSTRACT

Neutrophil extracellular trap formation (NETosis) and the NLR family pyrin domain containing 3 (NLRP3) inflammasome assembly are associated with a similar spectrum of human disorders. While NETosis is known to be regulated by peptidylarginine deiminase 4 (PAD4), the role of the NLRP3 inflammasome in NETosis was not addressed. Here, we establish that under sterile conditions the cannonical NLRP3 inflammasome participates in NETosis. We show apoptosis-associated speck-like protein containing a CARD (ASC) speck assembly and caspase-1 cleavage in stimulated mouse neutrophils without LPS priming. PAD4 was needed for optimal NLRP3 inflammasome assembly by regulating NLRP3 and ASC protein levels post-transcriptionally. Genetic ablation of NLRP3 signaling resulted in impaired NET formation, because NLRP3 supported both nuclear envelope and plasma membrane rupture. Pharmacological inhibition of NLRP3 in either mouse or human neutrophils also diminished NETosis. Finally, NLRP3 deficiency resulted in a lower density of NETs in thrombi produced by a stenosis-induced mouse model of deep vein thrombosis. Altogether, our results indicate a PAD4-dependent formation of the NLRP3 inflammasome in neutrophils and implicate NLRP3 in NETosis under noninfectious conditions in vitro and in vivo.


Subject(s)
Extracellular Traps/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Neutrophils/enzymology , Animals , Caspase 1/pharmacology , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Neutrophils/drug effects , Protein-Arginine Deiminase Type 4/metabolism , Venous Thrombosis/blood , Venous Thrombosis/enzymology , Venous Thrombosis/genetics
9.
J Neuroinflammation ; 18(1): 128, 2021 Jun 06.
Article in English | MEDLINE | ID: mdl-34092247

ABSTRACT

BACKGROUND: Patients with prior illness are more vulnerable to heat stroke-induced injury, but the underlying mechanism is unknown. Recent studies suggested that NLRP3 inflammasome played an important role in the pathophysiology of heat stroke. METHODS: In this study, we used a classic animal heat stroke model. Prior infection was mimicked by using lipopolysaccharide (LPS) or lipoteichoic acid (LTA) injection before heat stroke (LPS/LTA 1 mg/kg). Mice survival analysis curve and core temperature (TC) elevation curve were produced. NLRP3 inflammasome activation was measured by using real-time PCR and Western blot. Mice hypothalamus was dissected and neuroinflammation level was measured. To further demonstrate the role of NLRP3 inflammasome, Nlrp3 knockout mice were used. In addition, IL-1ß neutralizing antibody was injected to test potential therapeutic effect on heat stroke. RESULTS: Prior infection simulated by LPS/LTA injection resulted in latent inflammation status presented by high levels of cytokines in peripheral serum. However, LPS/LTA failed to cause any change in animal survival rate or body temperature. In the absence of LPS/LTA, heat treatment induced heat stroke and animal death without significant systemic or neuroinflammation. Despite a decreased level of IL-1ß in hypothalamus, Nlrp3 knockout mice demonstrated no survival advantage under mere heat exposure. In animals with prior infection, their heat tolerance was severely impaired and NLRP3 inflammasome induced neuroinflammation was detected. The use of Nlrp3 knockout mice enhanced heat tolerance and alleviated heat stroke-induced death by reducing mice hypothalamus IL-1ß production with prior infection condition. Furthermore, IL-1ß neutralizing antibody injection significantly extended endotoxemic mice survival under heat stroke. CONCLUSIONS: Based on the above results, NLRP3/IL-1ß induced neuroinflammation might be an important mechanistic factor in heat stroke pathology, especially with prior infection. IL-1ß may serve as a biomarker for heat stroke severity and potential therapeutic method.


Subject(s)
Brain/metabolism , Brain/pathology , Heat Stroke/complications , Heat Stroke/physiopathology , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuroinflammatory Diseases/complications , Neuroinflammatory Diseases/metabolism , Animals , Antibodies, Neutralizing/therapeutic use , Disease Models, Animal , Heat Stroke/drug therapy , Heat Stroke/pathology , Inflammasomes/metabolism , Interleukin-1beta/immunology , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Signal Transduction , Teichoic Acids , Thermotolerance
10.
J Immunol ; 206(10): 2441-2452, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33941658

ABSTRACT

Intestinal barrier is essential for dietary products and microbiota compartmentalization and therefore gut homeostasis. When this barrier is broken, cecal content overflows into the peritoneal cavity, leading to local and systemic robust inflammatory response, characterizing peritonitis and sepsis. It has been shown that IL-1ß contributes with inflammatory storm during peritonitis and sepsis and its inhibition has beneficial effects to the host. Therefore, we investigated the mechanisms underlying IL-1ß secretion using a widely adopted murine model of experimental peritonitis. The combined injection of sterile cecal content (SCC) and the gut commensal bacteria Bacteroides fragilis leads to IL-1ß-dependent peritonitis, which was mitigated in mice deficient in NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3) inflammasome components. Typically acting as a damage signal, SCC, but not B. fragilis, activates canonical pathway of NLRP3 promoting IL-1ß secretion in vitro and in vivo. Strikingly, absence of fiber in the SCC drastically reduces IL-1ß production, whereas high-fiber SCC conversely increases this response in an NLRP3-dependent manner. In addition, NLRP3 was also required for IL-1ß production induced by purified dietary fiber in primed macrophages. Extending to the in vivo context, IL-1ß-dependent peritonitis was worsened in mice injected with B. fragilis and high-fiber SCC, whereas zero-fiber SCC ameliorates the pathology. Corroborating with the proinflammatory role of dietary fiber, IL-1R-deficient mice were protected from peritonitis induced by B. fragilis and particulate bran. Overall, our study highlights a function, previously unknown, for dietary fibers in fueling peritonitis through NLRP3 activation and IL-1ß secretion outside the gut.


Subject(s)
Bacteroides Infections/immunology , Bacteroides fragilis/immunology , Dietary Fiber/adverse effects , Inflammasomes/metabolism , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Peritonitis/immunology , Animals , Bacteroides Infections/microbiology , Diet , Dietary Fiber/administration & dosage , Disease Models, Animal , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Peritonitis/microbiology , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/genetics , Signal Transduction/drug effects , Signal Transduction/immunology
11.
Front Immunol ; 12: 628168, 2021.
Article in English | MEDLINE | ID: mdl-33717152

ABSTRACT

Obstructive sleep apnea (OSA) associated neurocognitive impairment is mainly caused by chronic intermittent hypoxia (CIH)-triggered neuroinflammation and oxidative stress. Previous study has demonstrated that mitochondrial reactive oxygen species (mtROS) was pivotal for hypoxia-related tissue injury. As a cytosolic multiprotein complex that participates in various inflammatory and neurodegenerative diseases, NLRP3 inflammasome could be activated by mtROS and thereby affected by the mitochondria-selective autophagy. However, the role of NLRP3 and possible mitophagy mechanism in CIH-elicited neuroinflammation remain to be elucidated. Compared with wild-type mice, NLRP3 deficiency protected them from CIH-induced neuronal damage, as indicated by the restoration of fear-conditioning test results and amelioration of neuron apoptosis. In addition, NLRP3 knockout mice displayed the mitigated microglia activation that elicited by CIH, concomitantly with elimination of damaged mitochondria and reduction of oxidative stress levels (malondialdehyde and superoxide dismutase). Elevated LC3 and beclin1 expressions were remarkably observed in CIH group. In vitro experiments, intermittent hypoxia (IH) significantly facilitated mitophagy induction and NLRP3 inflammasome activation in microglial (BV2) cells. Moreover, IH enhanced the accumulation of damaged mitochondria, increased mitochondrial depolarization and augmented mtROS release. Consistently, NLRP3 deletion elicited a protective phenotype against IH through enhancement of Parkin-mediated mitophagy. Furthermore, Parkin deletion or pretreated with 3MA (autophagy inhibitor) exacerbated these detrimental actions of IH, which was accompanied with NLRP3 inflammasome activation. These results revealed NLRP3 deficiency acted as a protective promotor through enhancing Parkin-depended mitophagy in CIH-induced neuroinflammation. Thus, NLRP3 gene knockout or pharmacological blockage could be as a potential therapeutic strategy for OSA-associated neurocognitive impairment.


Subject(s)
Brain/metabolism , Inflammasomes/deficiency , Inflammation/prevention & control , Mitochondria/metabolism , Mitophagy , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Sleep Apnea Syndromes/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Brain/immunology , Brain/pathology , Cell Line , Disease Models, Animal , Hypoxia/complications , Inflammasomes/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , Microglia/metabolism , Microglia/pathology , Mitochondria/immunology , Mitochondria/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Neuroimmunomodulation , Oxidative Stress , Signal Transduction , Sleep Apnea Syndromes/immunology , Sleep Apnea Syndromes/pathology
12.
Toxicol Appl Pharmacol ; 418: 115495, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33741346

ABSTRACT

In the present study, the effects of NLRP3 on radiation-induced tissue damage, including colon and skin damage in mice, and the possible mechanisms were explored in vivo and in vitro. The mice were subjected to whole abdomen radiation by timed exposure to X-ray at a cumulative dose of 14 Gy. The survival rate showed that NLRP3 deficiency increased the mortality rate in mice. Furthermore, colon damage, evaluated by H&E staining and barrier function analysis, were significantly aggravated by NLRP3 deficiency. Enhanced phosphorylation of p-TBK1 and p-IRF3 in colonic tissue as well as elevated IFN-ß levels in the serum indicated hyperactivation of cGAS-STING signaling. Moreover, radiation-induced expression of p-TBK1, p-IRF3, and IFN-ß in BMDMs increased in vitro after NLRP3 knockout. Thus, our study outcomes suggest that NLRP3 may protect mice from radiation-induced tissue damage via attenuating cGAS-STING signaling.


Subject(s)
Colon/radiation effects , Macrophages/radiation effects , Membrane Proteins/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nucleotidyltransferases/metabolism , Radiation Injuries/prevention & control , Skin Ulcer/prevention & control , Skin/radiation effects , Animals , Cells, Cultured , Colon/enzymology , Colon/pathology , Interferon Regulatory Factor-3/metabolism , Interferon-beta/metabolism , Macrophages/enzymology , Macrophages/pathology , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Radiation Injuries/enzymology , Radiation Injuries/genetics , Radiation Injuries/pathology , Signal Transduction , Skin/enzymology , Skin/pathology , Skin Ulcer/enzymology , Skin Ulcer/genetics , Skin Ulcer/pathology
13.
Clin Sci (Lond) ; 135(5): 687-701, 2021 03 12.
Article in English | MEDLINE | ID: mdl-33620070

ABSTRACT

Muscle tissue damage is one of the local effects described in bothropic envenomations. Bothropstoxin-I (BthTX-I), from Bothrops jararacussu venom, is a K49-phospholipase A2 (PLA2) that induces a massive muscle tissue injury, and, consequently, local inflammatory reaction. The NLRP3 inflammasome is a sensor that triggers inflammation by activating caspase 1 and releasing interleukin (IL)-1ß and/or inducing pyroptotic cell death in response to tissue damage. We, therefore, aimed to address activation of NLRP3 inflammasome by BthTX-I-associated injury and the mechanism involved in this process. Intramuscular injection of BthTX-I results in infiltration of neutrophils and macrophages in gastrocnemius muscle, which is reduced in NLRP3- and Caspase-1-deficient mice. The in vitro IL-1ß production induced by BthTX-I in peritoneal macrophages (PMs) requires caspase 1/11, ASC and NLRP3 and is dependent on adenosine 5'-triphosphate (ATP)-induced K+ efflux and P2X7 receptor (P2X7R). BthTX-I induces a dramatic release of ATP from C2C12 myotubes, therefore representing the major mechanism for P2X7R-dependent inflammasome activation in macrophages. A similar result was obtained when human monocyte-derived macrophages (HMDMs) were treated with BthTX-I. These findings demonstrated the inflammatory effect of BthTX-I on muscle tissue, pointing out a role for the ATP released by damaged cells for the NLRP3 activation on macrophages, contributing to the understanding of the microenvironment of the tissue damage of the Bothrops envenomation.


Subject(s)
Crotalid Venoms/toxicity , Inflammasomes/metabolism , Inflammation/chemically induced , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Adenosine Triphosphate , Animals , Bothrops , Caspase 1/deficiency , Cell Line , Humans , Macrophages , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/pathology , Muscular Diseases/chemically induced , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Receptors, Purinergic P2X7
14.
Oxid Med Cell Longev ; 2021: 6679100, 2021.
Article in English | MEDLINE | ID: mdl-33628380

ABSTRACT

Inflammation has been considered a key component in the pathogenesis and progression of angiotensin II- (Ang II-) induced cardiac hypertrophy and related cardiomyopathy. As a vital mediator of inflammation, the role of the Nlrp3 inflammasome in Ang II-induced cardiomyopathy remains unclear. This study was aimed to determine whether Nlrp3 inflammasome activation and its downstream pathway were involved in Ang II-induced cardiomyopathy. We established an Ang II infusion model in both wild-type and Nlrp3-/- mice to determine the contribution of Nlrp3 to cardiac function. Cardiac fibrosis was determined by Masson's trichrome staining, real-time PCR, and TUNEL assay; cardiac function was assessed by echocardiography. Nlrp3 inflammasome activation and related downstream cytokines were measured by Western blotting and enzyme-linked immunosorbent assays; mitochondrial dysfunction was examined by transmission electron microscopy and real-time PCR. We found that Ang II-infused mice showed impaired cardiac function, as evidenced by increased cardiac fibrosis, apoptosis, inflammation, and left ventricular dysfunction. However, these alterations were significantly alleviated in the mice with Nlrp3 gene deletion. Moreover, Ang II-infused mice showed increased Nlrp3 inflammasome activity relative to that of the cytokines IL-1ß and IL-18, increased reactive oxygen species, mitochondrial abnormalities, and decreased mtDNA copy number and ATP synthase activity. These molecular and pathological alterations were also attenuated in Nlrp3 deficient mice. In conclusion, Nlrp3 inflammasome-induced mitochondrial dysfunction is involved in Ang II-induced cardiomyopathy. Nlrp3 gene deletion attenuated mitochondrial abnormalities, cardiac inflammation, oxidative stress, and fibrosis and thus alleviated heart dysfunction and hypertrophy. Targeting the Nlrp3 inflammasome and/or mitochondria may be a therapeutic approach for Ang II-induced cardiac diseases.


Subject(s)
Cardiomyopathies/metabolism , Cardiomyopathies/physiopathology , Mitochondria/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Angiotensin II , Animals , Apoptosis , Cardiomyopathies/chemically induced , Cardiomyopathies/diagnostic imaging , Cytokines/metabolism , Electrocardiography , Fibrosis , Gene Deletion , Inflammasomes/metabolism , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/ultrastructure , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress
15.
Neurochem Res ; 46(4): 770-777, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33400023

ABSTRACT

Intracerebral hemorrhage (ICH) is a devastating cerebrovascular disease with a high mortality rate affecting individuals worldwide. After ICH, persistent inflammation results in the death of brain cells, as well as the promotion of secondary brain injury. Verbascoside (VB), an active component in herbal medicine, possesses antioxidant, anti-inflammatory and neuroprotective properties. Furthermore, previous studies have shown that VB improves recovery of neuronal function after spinal cord injury in rats. In this study, we investigated whether VB limited inflammation induced by ICH through the targeting of NLRP3, which is associated with acute inflammation and apoptosis. Administration of VB reduced neurological impairment and pathological abnormalities associated with ICH, while increasing cell viability of neurons. This was achieved through NLRP3 inhibition and microglial activation. VB treatment decreased neuronal damage when co-cultured with microglia. Furthermore, knockout of NLRP3 eliminated the ability of VB to inhibit inflammation, cell death or protect neurons. Taken together, VB suppressed the inflammatory response following ICH by inhibiting NLRP3.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cerebral Hemorrhage/drug therapy , Glucosides/therapeutic use , Inflammation/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuroprotective Agents/therapeutic use , Phenols/therapeutic use , Animals , Apoptosis/drug effects , Cerebral Hemorrhage/complications , Dose-Response Relationship, Drug , Inflammation/etiology , Male , Mice, Inbred C57BL , Microglia/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency
16.
Mol Neurobiol ; 58(4): 1303-1311, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33169332

ABSTRACT

Recent researches showed that nucleotide-binding domain and leucine-rich repeat protein 3 (NLRP3) inflammasome inhibition exerted dopaminergic neuroprotection in cellular or animal models of Parkinson's disease (PD). NLRP3 inflammasome has been proposed as a drug target for treatment of PD. However, the interplay between chronic NLRP3 inflammasome and progressive α-synuclein pathology keeps poorly understood. Moreover, the potential mechanism keeps unknown. In the present study, we investigate whether NLRP3 inflammasome inhibition prevents α-synuclein pathology by relieving autophagy dysfunction in the chronic 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) mouse model of PD. NLRP3 knockout mice and their wild-type counterparts were treated with continuous MPTP administration via osmotic mini-pumps. Dopaminergic neuronal degeneration was assessed by western blotting and immunohistochemistry (IHC). The levels of dopamine and its metabolites were determined using high-performance liquid chromatography. NLRP3 inflammasome activation and autophagy biomarkers were assessed by western blot. The expressions of pro-inflammatory cytokines were measured by ELISA. The glial reaction and α-synuclein pathology were assessed by IHC and immunofluorescence. Our results show that NLRP3 inflammasome inhibition via NLRP3 knockout not only protects against nigral dopaminergic degeneration and striatal dopamine deletion but also prevents nigral pathological α-synuclein formation in PD mice. Furthermore, it significantly suppresses MPTP-induced glial reaction accompanied by the secretion of pro-inflammatory cytokines in the midbrain of mice. Most importantly, it relieves autophagy dysfunction in the midbrain of PD mice. Collectively, we demonstrate for the first time that improving autophagy function is involved in the preventive effect of NLRP3 inflammasome inhibition on α-synuclein pathology in PD.


Subject(s)
Autophagy , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , alpha-Synuclein/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Corpus Striatum/pathology , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Inflammation/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Nerve Degeneration/pathology , Protein Aggregates , Substantia Nigra/pathology
17.
Front Immunol ; 12: 765477, 2021.
Article in English | MEDLINE | ID: mdl-34987507

ABSTRACT

Nicotinamide adenine dinucleotide (NAD+) is an important cofactor in many redox and non-redox NAD+-consuming enzyme reactions. Intracellular NAD+ level steadily declines with age, but its role in the innate immune potential of myeloid cells remains elusive. In this study, we explored whether NAD+ depletion by FK866, a highly specific inhibitor of the NAD salvage pathway, can affect pattern recognition receptor-mediated responses in macrophages. NAD+-depleted mouse bone marrow-derived macrophages (BMDMs) exhibited similar levels of proinflammatory cytokine production in response to LPS or poly (I:C) stimulation compared with untreated cells. Instead, FK866 facilitated robust caspase-1 activation in BMDMs in the presence of NLRP3-activating signals such as ATP and nigericin, a potassium ionophore. However, this FK866-mediated caspase-1 activation was completely abolished in Nlrp3-deficient macrophages. FK866 plus nigericin stimulation caused an NLRP3-dependent assembly of inflammasome complex. In contrast, restoration of NAD+ level by supplementation with nicotinamide mononucleotide abrogated the FK866-mediated caspase-1 cleavage. FK866 did not induce or increase the expression levels of NLRP3 and interleukin (IL)-1ß but drove mitochondrial retrograde transport into the perinuclear region. FK866-nigericin-induced mitochondrial transport is critical for caspase-1 cleavage in macrophages. Consistent with the in vitro experiments, intradermal coinjection of FK866 and ATP resulted in robust IL-1ß expression and caspase-1 activation in the skin of wild-type, but not Nlrp3-deficient mice. Collectively, our data suggest that NAD+ depletion provides a non-transcriptional priming signal for NLRP3 activation via mitochondrial perinuclear clustering, and aging-associated NAD+ decline can trigger NLRP3 inflammasome activation in ATP-rich environments.


Subject(s)
Inflammasomes/immunology , NAD/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Animals , Cells, Cultured , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NAD/analysis , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency
18.
Pathog Dis ; 79(1)2021 01 09.
Article in English | MEDLINE | ID: mdl-33289808

ABSTRACT

A vast proportion of coronavirus disease 2019 (COVID-19) individuals remain asymptomatic and can shed severe acute respiratory syndrome (SARS-CoV) type 2 virus to transmit the infection, which also explains the exponential increase in the number of COVID-19 cases globally. Furthermore, the rate of recovery from clinical COVID-19 in certain pockets of the globe is surprisingly high. Based on published reports and available literature, here, we speculated a few immunovirological mechanisms as to why a vast majority of individuals remain asymptomatic similar to exotic animal (bats and pangolins) reservoirs that remain refractile to disease development despite carrying a huge load of diverse insidious viral species, and whether such evolutionary advantage would unveil therapeutic strategies against COVID-19 infection in humans. Understanding the unique mechanisms that exotic animal species employ to achieve viral control, as well as inflammatory regulation, appears to hold key clues to the development of therapeutic versatility against COVID-19.


Subject(s)
COVID-19/immunology , Cytokine Release Syndrome/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Receptors, KIR/immunology , Receptors, NK Cell Lectin-Like/immunology , Zoonoses/immunology , Animals , Animals, Exotic/virology , Asymptomatic Diseases , COVID-19/genetics , COVID-19/transmission , COVID-19/virology , Chiroptera/virology , Cytokine Release Syndrome/genetics , Cytokine Release Syndrome/prevention & control , Cytokine Release Syndrome/virology , Disease Reservoirs , Eutheria/virology , Gene Expression , Host Specificity , Humans , Immune Tolerance , Immunity, Innate , Interferon-beta/deficiency , Interferon-beta/genetics , Interferon-beta/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Monocytes/immunology , Monocytes/virology , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Receptors, KIR/deficiency , Receptors, KIR/genetics , Receptors, NK Cell Lectin-Like/deficiency , Receptors, NK Cell Lectin-Like/genetics , SARS-CoV-2/pathogenicity , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Zoonoses/genetics , Zoonoses/transmission , Zoonoses/virology
19.
FEBS J ; 288(10): 3186-3196, 2021 05.
Article in English | MEDLINE | ID: mdl-33145969

ABSTRACT

The release of interleukin (IL)-1ß from primary human monocytes in response to extracellular LPS occurs through the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome. In primary monocytes, in response to LPS, NLRP3 inflammasome activation is characterized by an independence of K+ efflux and ASC speck formation and has been termed the 'alternative' pathway. Here, we report that pharmacological inhibition of V-ATPase with bafilomycin A1 exacerbated LPS-induced NLRP3 inflammasome activation in primary human monocytes. Inhibition of V-ATPase in the presence of extracellular LPS led to NLRP3-dependent, K+ efflux-independent, ASC oligomerization and caspase-1 activation. Although V-ATPases are required for lysosomal acidification, we found that acidic lysosomal pH and protease activity were dispensable for this altered response, suggesting that V-ATPase inhibition triggered alternative signalling events. Therefore, V-ATPases may serve additional roles during NLRP3 inflammasome activation in primary human monocytes.


Subject(s)
Inflammasomes/drug effects , Lysosomes/drug effects , Macrolides/pharmacology , Monocytes/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Proton-Translocating ATPases/genetics , Caspase 1/genetics , Caspase 1/immunology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Humans , Hydrogen-Ion Concentration , Inflammasomes/immunology , Inflammasomes/metabolism , Inflammation , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Lipopolysaccharides/pharmacology , Lysosomes/immunology , Lysosomes/metabolism , Monocytes/cytology , Monocytes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/agonists , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Nigericin/pharmacology , Primary Cell Culture , Proton-Translocating ATPases/antagonists & inhibitors , Proton-Translocating ATPases/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Signal Transduction , THP-1 Cells
20.
Sci Rep ; 10(1): 21006, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33273482

ABSTRACT

Obesity-related diseases (e.g. type 2 diabetes mellitus and cardiovascular disorders) represent an increasing health problem worldwide. NLRP3 inflammasome activation may underlie obesity-induced inflammation and insulin resistance, and NLRP3 deficient mice exposed to high fat diet (HFD) appear to be protected from left ventricle (LV) concentric remodeling. Herein, we investigated if these beneficial effects were associated with alterations in plasma metabolites, using metabolomic and lipidomic analysis, and gut microbiota composition, using 16S rRNA sequencing of cecum content, comparing NLRP3 deficient and wild type (WT) mice on HFD and control diet. Obese NLRP3 deficient mice had lower systemic ceramide levels, potentially resulting attenuating inflammation, altered hepatic expression of fatty acids (FA) with lower mono-saturated FA and higher polyunsaturated FA levels, potentially counteracting development of liver steatosis, downregulated myocardial energy metabolism as assessed by proteomic analyses of LV heart tissue, and different levels of bile acids as compared with WT mice. These changes were accompanied by an altered composition of gut microbiota associated with decreased systemic levels of tri-methylamine-N-oxide and lipopolysaccharide, potentially inducing attenuating systemic inflammation and beneficial effects on lipid metabolism. Our findings support a role of NLRP3 inflammasome in the interface between metabolic and inflammatory stress, involving an altered gut microbiota composition.


Subject(s)
Diet, High-Fat/adverse effects , Gastrointestinal Microbiome , Metabolome , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Ceramides/blood , Energy Metabolism , Fatty Acids/blood , Fatty Acids/metabolism , Lipid Metabolism , Lipopolysaccharides/blood , Liver/metabolism , Male , Methylamines/blood , Mice , Mice, Inbred C57BL , Myocardium/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...