Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters










Publication year range
1.
Int J Biol Sci ; 18(2): 809-825, 2022.
Article in English | MEDLINE | ID: mdl-35002527

ABSTRACT

Diabetic keratopathy (DK) is an important diabetic complication at the ocular surface. Chronic low-grade inflammation mediated by the NLRP3 inflammasome promotes pathogenesis of diabetes and its complications. However, the effect of the NLRP3 inflammasome on DK pathogenesis remains elusive. Wild-type (WT) and Nlrp3 knockout (KO) C57 mice were used to establish a type I diabetes model by intraperitoneal injection of streptozotocin. The effect of the NLRP3 inflammasome on diabetic corneal wound healing and never regeneration was examined by a corneal epithelial abrasion model. Western blot, immunofluorescence staining, enzyme-linked immunosorbent assay (ELISA) and pharmacological treatment were performed to investigate the regulatory mechanism of advanced glycation end products (AGEs) on NLRP3 inflammasome activation and corneal wound healing in vivo. The cultured mouse corneal epithelial cells (TKE2) were used to evaluate the effect and mechanism of AGEs on NLRP3 inflammasome activation in vitro. We revealed that NLRP3 inflammasome-mediated inflammation and pyroptosis contributed to DK pathogenesis. Under physiological conditions, the NLRP3 inflammasome was required for corneal wound healing and nerve regeneration. However, under a diabetic scenario, sustained activation of the NLRP3 inflammasome resulted in postponed corneal wound healing and impaired nerve regeneration. Mechanistically, the accumulated AGEs promoted hyperactivation of the NLRP3 inflammasome through ROS production. Moreover, genetically and pharmacologically blocking the AGEs/ROS/NLRP3 inflammasome axis significantly expedited diabetic corneal epithelial wound closure and nerve regeneration. Our results revealed that AGEs-induced hyperactivation of the NLRP3 inflammasome resulted in delayed diabetic corneal wound healing and impaired nerve regeneration, which further highlighted the NLRP3 inflammasome as a promising target for DK treatment.


Subject(s)
Cornea/innervation , Corneal Diseases/genetics , Diabetes Mellitus, Experimental/complications , Glycation End Products, Advanced/administration & dosage , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Nerve Regeneration/genetics , Animals , Cornea/pathology , Corneal Diseases/etiology , Corneal Diseases/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/metabolism , Disease Models, Animal , Inflammasomes , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , Reactive Oxygen Species , Streptozocin , Wound Healing/drug effects , Wound Healing/genetics
2.
Life Sci ; 289: 120218, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34890588

ABSTRACT

BACKGROUND: Cancer and its therapies can impact fertility in various ways, and therefore a growing number of cancer survivors face fertility as a significant concern. The cytotoxic alkylating agent cyclophosphamide (CP) is commonly used as an antineoplastic agent; unfortunately, its use is significantly associated with male infertility and damage to the reproductive system. AIM: The present study aimed to assess the possible beneficial effects of Irbesartan (IRB) in a rat model of CP-induced testicular toxicity. MAIN METHODS: The effects of treatment were assessed by measuring peroxisome proliferator-activated receptor gamma (PPAR-γ) expression via qRT-PCR, the immunohistochemical (IHC) assessment of apoptotic markers, NOD-like receptor protein 3 (NLRP3), and nuclear factor-κB (NF-κB), determination of the count and viability of epididymal sperm, oxidative stress markers via biochemical analysis, serum testosterone, caspase-1, and interleukin-18 (IL-18) levels via ELISA, histopathological assessment, and fibrosis by Masson's trichrome (MT) stain. KEY FINDINGS: There was a significant increase in malondialdehyde (MDA), caspase-1, and IL-18 contents, NF-κB, NLRP3, Bcl-2-associated X protein (Bax), caspase-3, and MT staining in testicular tissue after CP administration compared to the normal control group. Whereas reduced glutathione (GSH), superoxide dismutase (SOD), PPAR-γ expression, B-cell lymphoma-2 (Bcl-2) staining, serum testosterone, and the count and viability of epididymal sperm were decreased compared to the normal control group. The IRB treatment has reversed CP-induced testicular toxicity. SIGNIFICANCE: It is possible to conclude that IRB revealed a significant testicular protective effect against CP via antioxidant, anti-apoptotic, and anti-inflammatory effects.


Subject(s)
Cyclophosphamide/adverse effects , Interleukin-18/biosynthesis , Irbesartan/pharmacology , NF-kappa B/biosynthesis , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , PPAR gamma/biosynthesis , Signal Transduction/drug effects , Testicular Diseases , Up-Regulation/drug effects , Animals , Cyclophosphamide/pharmacology , Infertility, Male/chemically induced , Infertility, Male/drug therapy , Infertility, Male/metabolism , Infertility, Male/pathology , Male , Rats , Testicular Diseases/chemically induced , Testicular Diseases/drug therapy , Testicular Diseases/metabolism , Testicular Diseases/pathology
3.
Front Immunol ; 12: 753287, 2021.
Article in English | MEDLINE | ID: mdl-34777363

ABSTRACT

Microbiota can exert immunomodulatory effects by short-chain fatty acids (SCFA) in experimental models of graft-versus-host disease (GvHD) after allogeneic hematopoietic stem cell transplantation (allo-SCT). Therefore we aimed to analyze the expression of SCFAs sensing G-protein coupled receptor GPR109A and GPR43 by quantitative PCR in 338 gastrointestinal (GI) biopsies obtained from 199 adult patients undergoing allo-SCT and assessed the interaction of GPR with FOXP3 expression and regulatory T cell infiltrates. GPR expression was strongly upregulated in patients with stage II-IV GvHD (p=0.000 for GPR109A, p=0.01 for GPR43) and at the onset of GvHD (p 0.000 for GPR109A, p=0.006 for GPR43) and correlated strongly with FOXP3 and NLRP3 expression. The use of broad-spectrum antibiotics (Abx) drastically suppressed GPR expression as well as FOXP3 expression in patients' gut biopsies (p=0.000 for GPRs, FOXP3 mRNA and FOXP3+ cellular infiltrates). Logistic regression analysis revealed treatment with Abx as an independent factor associated with GPR and FOXP3 loss. The upregulation of GPRs was evident only in the absence of Abx (p=0.001 for GPR109A, p=0.014 for GPR43) at GvHD onset. Thus, GPR expression seems to be upregulated in the presence of commensal bacteria and associates with infiltration of FOXP3+ T regs, suggesting a protective, regenerative immunomodulatory response. However, Abx, which has been shown to induce dysbiosis, interferes with this protective response.


Subject(s)
Anti-Bacterial Agents/adverse effects , Dysbiosis/chemically induced , Gastrointestinal Microbiome/drug effects , Graft vs Host Disease/microbiology , Intestines/metabolism , Receptors, Cell Surface/biosynthesis , Receptors, G-Protein-Coupled/biosynthesis , Adult , Allografts , Anti-Bacterial Agents/pharmacology , Biopsy , Butyrates/pharmacology , Cell Line, Tumor , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dysbiosis/microbiology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fatty Acids, Volatile/physiology , Female , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Graft vs Host Disease/metabolism , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Immunomodulation , Intestines/microbiology , Intestines/pathology , Male , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled/genetics , Severity of Illness Index , Symbiosis , T-Lymphocytes, Regulatory/immunology , Up-Regulation
4.
J Neuroimmunol ; 360: 577707, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34507013

ABSTRACT

Intrauterine inflammation affects fetal development of the nervous system and may cause prenatal brain injury in offspring. Previously, neural stem cells have been extensively used as a therapeutic choice for nervous system diseases. Recently, the therapeutic ability of conditioned medium, harvested from cultured stem cells, has captured the attention of researchers in the field. Our study aimed to compare the therapeutic effect of neural stem cells (NSCs) or NSC-conditioned medium (NSC-CM) after prenatal brain injury. The animal model was induced by intraperitoneal injection of lipopolysaccharide into the pregnant mice and NSCs or NSC-CM were transplanted into the lateral ventricle of embryos in treatment groups. Inflammation and apoptosis were evaluated postpartum in offspring via measuring the expression of NLRP3 gene and protein, the expression and the activity of caspase-3, and the expression of pro-inflammatory cytokines by real-time PCR, immunohistochemistry, western blotting, ELISA, and colorimetric assay kit. A rotarod test was performed for motor function evaluation. Data showed that although NSC-CM fought against the inflammation and apoptosis and improved the motor function, NSCs acted more efficiently. In conclusion, the results of our study contend that NSCs have a better therapeutic effect than CM in prenatal brain injury.


Subject(s)
Brain Injuries/therapy , Culture Media, Conditioned/pharmacology , Encephalitis/therapy , Fetal Diseases/therapy , Fetal Therapies , Neural Stem Cells/transplantation , Animals , Apoptosis , Brain Injuries/embryology , Brain Injuries/etiology , Caspase 3/biosynthesis , Caspase 3/genetics , Cells, Cultured , Cytokines/biosynthesis , Cytokines/genetics , Disease Models, Animal , Encephalitis/etiology , Female , Fetal Diseases/etiology , Inflammasomes/physiology , Injections, Intraperitoneal , Injections, Intraventricular , Lateral Ventricles , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/toxicity , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Pregnancy , Prenatal Exposure Delayed Effects , Rotarod Performance Test
5.
Am J Physiol Lung Cell Mol Physiol ; 321(5): L803-L813, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34431396

ABSTRACT

Chaperonin 60.1 (Cpn60.1) is a protein derived from Mycobacterium tuberculosis that has been shown, along with its peptide fragment IRL201104, to have beneficial effects in models of allergic inflammation. To further investigate the anti-inflammatory properties of Cpn60.1 and IRL201104, we have investigated these molecules in a model of nonallergic lung inflammation. Mice were treated with Cpn60.1 (0.5-5,000 ng/kg) or IRL201104 (0.00025-2.5 ng/kg), immediately before intranasal instillation of bacterial lipopolysaccharide (LPS). Cytokine levels and cell numbers in mouse bronchoalveolar lavage (BAL) fluid were measured 4 h after LPS administration. In some experiments, mice were depleted of lung-resident phagocytes. Cells from BAL fluid were analyzed for inflammasome function. Human umbilical vein endothelial cells (HUVECs) were analyzed for adhesion molecule expression. Human neutrophils were analyzed for integrin expression, chemotaxis, and cell polarization. Cpn60.1 and IRL201104 significantly inhibited neutrophil migration into the airways, independently of route of administration. This effect of the peptide was absent in TLR4 and annexin A1 knockout mice. Intravital microscopy revealed that IRL201104 reduced leukocyte adhesion and migration into inflamed tissues. However, IRL201104 did not significantly affect adhesion molecule expression in HUVECs or integrin expression, chemotaxis, or polarization of human neutrophils at the studied concentrations. In phagocyte-depleted animals, the anti-inflammatory effect of IRL201104 was not significant. IRL201104 significantly reduced IL-1ß and NLRP3 expression and increased A20 expression in BAL cells. This study shows that Cpn60.1 and IRL201104 potently inhibit LPS-induced neutrophil infiltration in mouse lungs by a mechanism dependent on tissue-resident phagocytes and to a much lesser extent, the proresolving factor annexin A1.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chaperonin 60/pharmacology , Chaperonins/pharmacology , Neutrophil Infiltration/drug effects , Peptide Fragments/pharmacology , Pneumonia/prevention & control , Animals , Annexin A1/genetics , Bronchoalveolar Lavage Fluid/chemistry , Cell Adhesion/drug effects , Cell Movement/drug effects , Cells, Cultured , Cytokines/analysis , Female , Human Umbilical Vein Endothelial Cells , Humans , Integrins/biosynthesis , Interleukin-1beta/biosynthesis , Lipopolysaccharides/toxicity , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , Neutrophils/immunology , Toll-Like Receptor 4/genetics
6.
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
7.
Mediators Inflamm ; 2021: 5551578, 2021.
Article in English | MEDLINE | ID: mdl-34093086

ABSTRACT

BACKGROUND: The functional characteristics of NLRP3 in the pathogenesis of coxsackievirus B3- (CVB3-) induced viral myocarditis (VMC) have not been fully elucidated, and the targeted therapeutic effect of NLRP3 or its related pathway in VMC has not been reported. METHOD: In this work, the change patterns of NLRP3- and Th17-related factors were detected during the pathological process of CVB3-induced VMC in Balb/c mice. The correlation between NLRP3 and Th17 cells during the VMC process was analyzed by Spearman test. The coculture system of spleen CD4+ T and bone marrow CD11c+ DC cells was set to explore the orchestration of NLRP3 and Th17 in the pathological development of VMC in vitro. Anti-IL-1ß antibody or NLRP3-/- Balb/c were used to block the NLRP3 pathway indirectly and directly to analyze the NLRP3-targeting therapeutic value. RESULTS: The change patterns of NLRP3- and Th17-related molecules in the whole pathological process of mouse CVB3-induced VMC were described. Through Spearman correlation analysis, it was confirmed that there was a close correlation between NLRP3 and Th17 cells in the whole pathological process of VMC. And the interaction mode between NLRP3 and Th17 was preliminarily explored in the cell experiment in vitro. Under the intervention of an anti-IL-1ß antibody or NLRP3 knockout, the survival rate of the intervention group was significantly improved, the degree of myocardial inflammation and fibrosis was significantly alleviated, and the content of myocardial IL-17 and spleen Th17 was also significantly decreased. CONCLUSION: Our findings demonstrated a key role of the NLRP3 inflammasome and its close relationship with Th17 in the pathological progression of CVB3-induced VMC and suggested a possible positive feedback-like mutual regulation mechanism between the NLRP3 inflammasome and Th17 in vitro and in the early stage of CVB3 infection. Taking NLRP3 as a new starting point, it provides a new target and idea for the prevention and treatment of CVB3-induced VMC.


Subject(s)
Coxsackievirus Infections/drug therapy , Coxsackievirus Infections/virology , Enterovirus B, Human , Myocarditis/drug therapy , Myocarditis/therapy , Myocarditis/virology , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , Th17 Cells/cytology , Animals , CD11c Antigen/biosynthesis , CD4-Positive T-Lymphocytes/cytology , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Genotype , Humans , Immunohistochemistry , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Laryngeal Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Myocarditis/immunology , Spleen/metabolism
8.
Exp Eye Res ; 209: 108668, 2021 08.
Article in English | MEDLINE | ID: mdl-34144035

ABSTRACT

Vitamin D (VD) deficiency delays corneal wound healing in those with diabetes, which cannot be rescued with supplemental diet. Here, we employed topical calcitriol application to evaluate its efficiency in corneal wound healing and reinnervation in diabetic mice. Type 1 diabetic mice were topically administrated calcitriol, or subconjunctivally injected with NLRP3 antagonist MCC950 or IL-1ß blocking antibody after epithelial debridement. Serum VD levels, corneal epithelial defect, corneal sensation and nerve density, NLRP3 inflammasome activation, neutrophil infiltration, macrophage phenotypes, and gene expressions were examined. Compared with those of normal mice, diabetic mice showed reduced serum VD levels. Topical calcitriol application promoted corneal wound healing and nerve regeneration, as well as sensation recovery in diabetic mice. Moreover, calcitriol ameliorated neutrophil infiltration and promoted the M1-to-M2 macrophage transition, accompanied by suppressed overactivation of the NLRP3 inflammasome. Treatment with NLRP3 antagonist or IL-1ß blockage demonstrated similar improvements as those of topical calcitriol application. Additionally, calcitriol administration upregulated desmosomal and hemidesmosomal gene expression in the diabetic cornea. In conclusion, topical calcitriol application promotes corneal wound healing and reinnervation during diabetes, which may be related to the suppression of the overactivation of NLRP3 inflammasome.


Subject(s)
Calcitriol/administration & dosage , Cornea/innervation , Corneal Diseases/genetics , Diabetes Mellitus, Experimental/complications , Gene Expression Regulation , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Nerve Regeneration/genetics , Animals , Cornea/pathology , Corneal Diseases/etiology , Corneal Diseases/metabolism , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal , Inflammasomes , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , RNA/genetics , Wound Healing/drug effects , Wound Healing/genetics
9.
J Chem Neuroanat ; 116: 101994, 2021 10.
Article in English | MEDLINE | ID: mdl-34166779

ABSTRACT

Hypoxia inducible factor 1 alpha (HIF-1α) is involved in regulating the biological functions of neuronal death after traumatic brain injury (TBI), and attaches importance in the inflammatory response, but its potential mechanism is still unknown. Our study aimed to explore the regulatory mechanism between HIF-1α and NLRP3 inflammasome after TBI. Male mice underwent controlled cortical impact (CCI) or sham-operated procedures. Brain water content and blood-brain barrier permeability were measured at the indicated time after TBI. The behavioral performance, ELISA, immunofluorescence, and western blot analysis were used to determine whether HIF-1α specifically targeted TBI-induced pyroptosis. We discovered that TBI-induced brain injury caused by external mechanical forces is characterized by edema and blood-brain barrier disorder, and the release of IL-1ß, IL-18, and LDH and upregulation of HIF-1α expression, reaching the peak on the third day post-TBI. In addition, HIF-1α accumulated NLRP3 inflammasome-mediated pyroptosis and activation of microglia. The protein expressions of NLRP3, GSDMD, GSDMD-N, pro-caspase 1, and cleaved caspase 1 were markedly increased in the injured cortex, which were restored to normal levels by the interference of HIF-1α. The inactivation of HIF-1α conferred neuroprotection and alleviated brain injury after TBI. HIF-1α was implicated in TBI-induced brain injury, aggravated NLRP3 inflammasome -mediated pyroptosis, and the activation of microglia, which provided a potential target for treating TBI.


Subject(s)
Brain Injuries, Traumatic/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Inflammasomes/biosynthesis , Microglia/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , Pyroptosis/physiology , Animals , Brain Injuries, Traumatic/pathology , Cell Line , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/pathology
10.
Exp Eye Res ; 209: 108654, 2021 08.
Article in English | MEDLINE | ID: mdl-34102208

ABSTRACT

Diabetes mellitus (DM) induces damage to the ocular surface, which leads to vision decline. In the current study, we investigated whether N-acetylcysteine (NAC) plays a protective role in diabetes-induced ocular surface damage. The diabetic mice model was treated with 0.3% NAC topically. Corneal epithelial integrity, tear volume and corneal sensitivity were examined by sodium fluorescein staining, phenol red cotton thread and esthesiometer respectively. The level of reactive oxygen species (ROS) was measured with 2',7-dichlorofluorescein diacetate. The expression of NLRP3, IL-1ß and caspase-1 were evaluated by RT-PCR, western blot and immunostaining. The level of SOD1 was assessed by RT-PCR. We found that the expression of NLRP3, IL-1ß and caspase-1 were elevated in diabetic cornea and conjunctiva. Treatment with NAC improved corneal epithelial integrity, increased tear production and corneal sensitivity in diabetic mice. Moreover, NAC markedly attenuated ROS accumulation and decreased NLRP3, IL-1ß and caspase-1 levels in diabetic cornea and conjunctiva. These results suggest that NAC improves ocular surface damage in STZ-induced diabetic mice, which may be related to the inhibition of the ROS/NLRP3/Caspase-1/IL-1ß signaling pathway.


Subject(s)
Caspase 1/genetics , Conjunctiva/metabolism , Cornea/metabolism , Diabetes Mellitus, Experimental/genetics , Gene Expression Regulation , Interleukin-1beta/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Animals , Blotting, Western , Caspase 1/biosynthesis , Conjunctiva/pathology , Cornea/pathology , Diabetes Mellitus, Experimental/metabolism , Inflammasomes/genetics , Inflammasomes/metabolism , Interleukin-1beta/biosynthesis , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , RNA/genetics , Reactive Oxygen Species/metabolism , Signal Transduction
11.
Neurobiol Dis ; 156: 105399, 2021 08.
Article in English | MEDLINE | ID: mdl-34029695

ABSTRACT

Immune system hypersensitivity is believed to contribute to mental frailty in the elderly. Solid evidence indicates NOD-like receptor pyrin domain containing-3 (NLRP3)-inflammasome activation intimately connects aging-associated chronic inflammation (inflammaging) to senile cognitive decline. Thioredoxin interacting protein (TXNIP), an inducible protein involved in oxidative stress, is essential for NLRP3 inflammasome activity. This study aims to find whether TXNIP/NLRP3 inflammasome pathway is involved in senile dementia. According to our studies on sex-matched mice, TXNIP was significantly upregulated in aged animals, paralleled by the NLRP3-inflammasome over-activity leading to enhanced caspase-1 cleavage and IL-1ß maturation, in both sexes. This was closely associated with depletion of the anti-aging and cognition enhancing protein klotho, in aged males. Txnip knockout reversed age-related NLRP3-hyperactivity and enhanced thioredoxin (TRX) levels. Further, TXNIP inhibition along with verapamil replicated TXNIP/NLRP3-inflammasome downregulation in aged animals, with FOXO-1 and mTOR upregulation. These alterations concurred with substantial improvements in both cognitive and sensorimotor abilities. Together, these findings substantiate the pivotal role of TXNIP to drive inflammaging in parallel with klotho depletion and functional decline, and delineate thioredoxin system as a potential target to decelerate senile dementia.


Subject(s)
Aging/metabolism , Brain/metabolism , Carrier Proteins/biosynthesis , Inflammation Mediators/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , Thioredoxins/biosynthesis , Aging/genetics , Aging/pathology , Animals , Brain/pathology , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Female , Inflammation Mediators/antagonists & inhibitors , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Oxidative Stress/physiology , Thioredoxins/antagonists & inhibitors , Thioredoxins/genetics
12.
Exp Eye Res ; 209: 108640, 2021 08.
Article in English | MEDLINE | ID: mdl-34058229

ABSTRACT

Diabetic retinopathy (DR) has been considered to involve mitochondrial alterations and be related to the nucleotide-binding oligomerization domain-like receptors 3 (NLRP3) inflammasome activation. The voltage-dependent anion channel 1 (VDAC1) protein is one of the key proteins that regulates the metabolic and energetic functions of the mitochondria. To explore the involvement of VDAC1 in mitophagy regulation of NLRP3 inflammasome activation under high-glucose (HG) conditions, this study examined expressions of VDAC1, mitochondrial function and mitophagy-related proteins, and NLRP3 inflammasome-related proteins in human retinal capillary endothelial cells (HRCECs) cultured with 30 mM of glucose in the presence or absence of mitophagy inhibitor (Mdivi-1) using Western blot. Mitochondrial membrane potential and mitochondrial reactive oxygen species (mtROS) were detected using flow cytometry. GFP-tagged pAdTrack-VDAC1 adenovirus was used to overexpress VDAC1. Cell biological behaviors, including proliferation, migration, tubule formation, and apoptosis, were also observed. Our results showed that when compared to the normal glucose and high mannitol groups, increased amounts of mitochondrial fragments, reduced mitochondrial membrane potential, increased expression of mitochondrial fission protein Drp 1, decreased expression of mitochondrial fusion protein Mfn 2, accumulation of mtROS, and activation of the NLRP3 inflammasome were observed in the HG group. Meanwhile, HG markedly reduced the protein expressions of PINK1, Parkin and VDAC1. Inhibition of mitophagy reduced PINK1 expression, enhanced NLRP3 expression, but failed to alter VDAC1. VDAC1 overexpression promoted PINK1 expression, inhibited NLRP3 activation and changed the cell biological behaviors under HG conditions. These findings demonstrate that VDAC1-mediated mitophagy plays a crucial role in regulating NLRP3 inflammasome activation in retinal capillary endothelial cells under HG conditions, suggesting that VDAC1 may be a potential target for preventing or treating DR.


Subject(s)
Endothelial Cells/metabolism , Gene Expression Regulation , Inflammasomes/genetics , Mitophagy/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Voltage-Dependent Anion Channel 1/biosynthesis , Voltage-Dependent Anion Channel 1/genetics , Apoptosis , Capillaries/metabolism , Capillaries/ultrastructure , Cells, Cultured , Endothelial Cells/ultrastructure , Endothelium, Vascular/metabolism , Endothelium, Vascular/ultrastructure , Humans , Inflammasomes/metabolism , Membrane Potential, Mitochondrial/physiology , Microscopy, Electron, Transmission , Mitochondria/metabolism , Mitochondria/ultrastructure , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , RNA/genetics , Retinal Vessels/metabolism , Retinal Vessels/ultrastructure
13.
Exp Eye Res ; 208: 108595, 2021 07.
Article in English | MEDLINE | ID: mdl-34000276

ABSTRACT

This study aimed to explore the effects of N-acetylserotonin (NAS) on the expression of interleukin-1ß (IL-1ß) in the retina of retinal ischemia-reperfusion injury (RIRI) rats via the toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-κB)/nod-like receptor pyrin domain containing 3 (NLRP3) signaling pathway. In this study, adult male Sprague Dawley rats were randomly divided into the sham, RIRI, RIRI + NAS and RIRI + TAK-242 + NAS groups. The rats in the RIRI + NAS and RIRI + TAK-242 + NAS groups were intraperitoneally injected with NAS 30 min before and after modeling. TAK-242, a selective TLR4 inhibitor, was administered by intraperitoneal injection in RIRI + TAK-242 + NAS group. The RIRI rat model was established by elevating the intraocular pressure to 110 mmHg for 60 min. The retinal structure and edema were assessed by H&E staining. The expression levels of TLR4, phosphorylated NF-κB (p-NF-κB), NLRP3, cleaved Caspase-1, and IL-1ß in the retina of each group were detected using immunohistochemistry and Western blot. The correlations of the differences of TLR4+ and cleaved Caspase-1+ with IL-1ß+ cells (between the NAS and the RIRI groups) were analyzed, using linear regression in the RIRI + NAS group. Results showed that thinner retina, more RGCs, and less TLR4+, p-NF-κB+, NLRP3+, cleaved Caspase-1+, and IL-1ß+ cells in the retina were observed in the RIRI + NAS and RIRI + TAK-242 + NAS groups compared with the RIRI group 12 h after RIRI (all P < 0.01). Western blot analysis results showed that the expression of IL-1ß in the RIRI + NAS group began to increase 6 h after RIRI, and it reached a high level 12 h after RIRI, and then decreased. And it was lower at each time point in the RIRI + NAS group than in the RIRI group, and there existed significant difference (all P < 0.01). Besides, the expression levels of TLR4, p-NF-κB, NLRP3, and cleaved Caspase-1 proteins in the RIRI + NAS and RIRI + TAK-242 + NAS groups decreased 12 h after RIRI compared with those in the RIRI group (all P < 0.01). The difference in IL-1ß+ cells was significantly correlated with those of TLR4+ and cleaved Caspase-1+ cells in the RIRI + NAS group (r2 = 0.9054 or 0.7431, P < 0.01). In conclusion, NAS could attenuate the expression of IL-1ß by inhibiting the TLR4/NF-κB/NLRP3 signaling pathway, reduce the retina edema, and promote the survival of RGCs, thereby alleviating the retinal injury and exert its neuroprotective effect.


Subject(s)
Interleukin-18/biosynthesis , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , Reperfusion Injury/metabolism , Retinal Diseases/metabolism , Serotonin/analogs & derivatives , Toll-Like Receptor 4/biosynthesis , Animals , Disease Models, Animal , Immunohistochemistry , Inflammasomes/metabolism , Male , Rats , Rats, Sprague-Dawley , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Retinal Diseases/drug therapy , Retinal Diseases/pathology , Serotonin/pharmacology , Signal Transduction/drug effects
14.
Neurol Res ; 43(8): 668-676, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33829970

ABSTRACT

Objectives: Stroke is an important cause of death and disability. Recent evidence suggests that post-stroke inflammation is an important factor in stroke pathology and a root cause of its lasting consequences. Phenothiazine drugs, like chlorpromazine and promethazine (C + P), induce hypothermia and have been shown to play a major role in neuroprotection. In the present study, we investigated this neuroprotective mechanism by assessing the anti-inflammatory effect of these drugs.Methods: Adult Sprague-Dawley rats underwent 2 h of middle cerebral artery occlusion (MCAO) followed by 6 or 24 h of reperfusion, with or without C + P (8 mg/kg). Infarct volumes, neurological deficits, along with mRNA and protein quantities of receptor-interacting protein 1 (RIP1), receptor-interacting protein 3 (RIP3), NLRPyrin domain containing 3 (NLRP3), and interleukin-1ß (IL-1ß) were assessed, as well as the infiltration of neutrophils and macrophages.Results: C + P induced hypothermia that significantly reduced RIP1, RIP3, NLRP3 and IL-1ß expression, infarction, and immune cell infiltration, while C + P treatment with temperature control at 37°C induced lesser effect.Conclusion: These findings suggest that the anti-inflammatory effect of C + P may be dependent on drug-induced hypothermia and regulation of the NLRP3 inflammasome via the RIP1/RIP3 complex. Future investigations are needed regarding C + P as potential treatment of ischemic stroke.


Subject(s)
Brain Injuries/drug therapy , Chlorpromazine/administration & dosage , Ischemic Stroke/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Promethazine/administration & dosage , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Brain Injuries/metabolism , Brain Injuries/pathology , Drug Therapy, Combination , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Male , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , Rats , Rats, Sprague-Dawley , Receptor-Interacting Protein Serine-Threonine Kinases/biosynthesis
15.
Respir Res ; 22(1): 45, 2021 Feb 06.
Article in English | MEDLINE | ID: mdl-33549095

ABSTRACT

BACKGROUND: S100 calcium binding protein A9 (S100A9) is a pro-inflammatory alarmin associated with several inflammation-related diseases. However, the role of S100A9 in lung injury in sepsis has not been fully investigated. Therefore, the present study aimed to determine the role of S100A9 in a lipopolysaccharide (LPS)-induced lung injury murine model and its underlying molecular mechanisms. METHODS: LPS was utilized to induce sepsis and lung injury in C57BL/6 or NOD-like receptor family pyrin domain containing 3 (NLRP3)-/- mice. To investigate the effects of S100A9 blockade, mice were treated with a specific inhibitor of S100A9. Subsequently, lung injury and inflammation were evaluated by histology and enzyme­linked immunosorbent assay (ELISA), respectively. Furthermore, western blot analysis and RT-qPCR were carried out to investigate the molecular mechanisms underlying the effects of S100A9. RESULTS: S100A9 was upregulated in the lung tissues of LPS-treated mice. However, inhibition of S100A9 alleviated LPS-induced lung injury. Additionally, S100A9 blockade also attenuated the inflammatory responses and apoptosis in the lungs of LPS-challenged mice. Furthermore, the increased expression of NLRP3 was also suppressed by S100A9 blockade, while S100A9 blockade had no effect on NLRP3-/- mice. In vitro, S100A9 downregulation mitigated LPS-induced inflammation. Interestingly, these effects were blunted by NLRP3 overexpression. CONCLUSION: The results of the current study suggested that inhibition of S100A9 could protect against LPS-induced lung injury via inhibiting the NLRP3 pathway. Therefore, S100A9 blockade could be considered as a novel therapeutic strategy for lung injury in sepsis.


Subject(s)
Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Calgranulin B/biosynthesis , Lipopolysaccharides/toxicity , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , Acute Lung Injury/prevention & control , Animals , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
16.
Inflammation ; 44(4): 1370-1380, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33630211

ABSTRACT

L-arginine (Arg), a semi-essential amino acid, has recently been shown to attenuate inflammatory response during cardiovascular disease. NLRP3 inflammasome serves a central role in amplification of cellular inflammation. In this study, we aimed to confirm the modulatory effect of Arg on NLRP3 inflammasome and the underlying mechanisms in vascular endothelial cells (ECs). Arg suppressed NLRP3 inflammasome activation in ECs stimulated with lipopolysaccharide (LPS) and adenosine triphosphate (ATP). Moreover, treatment with Arg increased the expression of the deacetylase sirtuin 1 (SIRT1) in ECs. Importantly, knockdown of SIRT1 abolished the inhibitory potential of Arg on the activation of NLRP3 inflammasome. Further study indicated that Arg also alleviated LPS plus ATP-induced the generation of reactive oxygen species (ROS) in ECs. In addition, Arg may regulate NLRP3 inflammasome activation partly through suppression of ROS production. In combination, we speculate that Arg exerts an inhibitory effect on the activation of NLRP3 inflammasome in ECs, which may be partly mediated by SIRT1 and ROS.


Subject(s)
Arginine/pharmacology , Endothelium, Vascular/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , Sirtuin 1/biosynthesis , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Lipopolysaccharides/toxicity , Molecular Docking Simulation , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Sirtuin 1/antagonists & inhibitors
17.
Cell Cycle ; 20(3): 332-344, 2021 02.
Article in English | MEDLINE | ID: mdl-33467979

ABSTRACT

This study aimed to determine the mechanism underlying the regulation of gout by the HOX transcript antisense RNA (HOTAIR) long non-coding RNA (lncRNA). The expression levels of HOTAIR, miR-20b, and Nlrp3 were estimated by qRT-PCR and western blotting. The methylation level of HOTAIR was detected by methylation-specific PCR. The recruitment of DNA methyltransferase 1 (DNMT1) to the lncRNA HOTAIR promoter was confirmed by a ChIP assay. RNA immunoprecipitation and RNA pull-down assays were used to confirm the interaction between HOTAIR and miR-20b. LncRNA HOTAIR and Nlrp3 expression was upregulated, and that of miR-20b was downregulated in synovial fluid mononuclear cells (SFMCs) collected from patients with gouty arthritis and monosodium urate (MSU)-stimulated THP-1 cells. Interleukin (IL)-1ß level increased substantially upon stimulation by MSU crystals. The methylation percentage of HOTAIR was reduced in SFMCs from patients with gouty arthritis and MSU-stimulated THP-1 cells. DNMT1 expression was downregulated in MSU-stimulated THP-1 cells, and DNMT1 knockdown increased lncRNA HOTAIR expression. In addition, the interaction of HOTAIR with miR-20b was confirmed. HOTAIR knockdown suppressed Nlrp3 expression and the secretion of inflammatory cytokines via miR-20b regulation. Finally, in vivo experiments showed that HOTAIR knockdown alleviated ankle swelling in a mouse model of gouty arthritis. These findings suggest that lncRNA HOTAIR knockdown suppresses inflammatory cytokine secretion by upregulating miR-20b and downregulating NLRP3, thereby alleviating ankle swelling in gouty arthritis.


Subject(s)
Arthritis, Gouty/metabolism , Down-Regulation/physiology , MicroRNAs/biosynthesis , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , RNA, Long Noncoding/biosynthesis , Up-Regulation/physiology , Aged , Animals , Arthritis, Gouty/genetics , Female , Gene Knockdown Techniques/methods , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Middle Aged , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , THP-1 Cells
18.
J Mol Med (Berl) ; 99(3): 403-414, 2021 03.
Article in English | MEDLINE | ID: mdl-33409553

ABSTRACT

Acute myeloid leukemia (AML) is an aggressive hematologic malignancy with poor prognosis and overall survival. Clinical investigations show that chronic stress is commonly present in the course of AML and associated with adverse outcome. However, the underlying molecular mechanisms are elusive. In the present study, a chronic restraint stress mouse model was established to evaluate the effect of stress on AML. We found that mice under chronic stress exhibited significantly increased liver and spleen infiltration of leukemic cells and poorer overall survival. This was accompanied by elevated cellular NLR family pyrin domain containing 3 (NLRP3) and interleukin-1ß (IL-1ß) in the liver or bone marrow, and secreted IL-1ß in the plasma, indicating the activation of inflammasomes under chronic restraint stress. High mobility group box 1 (HMGB1) expression was markedly increased in newly diagnosed AML patients, but reduced in complete remission AML patients. The expression level of HMGB1 was positively correlated with NLRP3 mRNA in AML patients. Knockdown of HMGB1 significantly decreased NLRP3 and IL-1ß expression in AML cell lines, and secreted IL-1ß in supernatant of AML cell culture, while HMGB1 stimulation caused contrary effects. These results implied that HMGB1 could be involved in the regulation of inflammasome activation in AML development. Mice model showed that chronic restraint stress-facilitated proliferation and infiltration of AML cells were largely abrogated by knocking down HMGB1. Knockdown of HMGB1 also ameliorated overall survival and remarkably neutralized NLRP3 and IL-1ß expression under chronic restraint stress. These findings provide evidences that chronic stress promotes AML progression via HMGB1/NLRP3/IL-1ß dependent mechanism, suggesting that HMGB1 is a potential therapeutic target for AML. KEY MESSAGES: • Chronic restraint stress promoted acute myeloid leukemia (AML) progression and mediated NLRP3 inflammasome activation in xenograft mice. • HMGB1 mediated NLRP3 inflammasome activation in AML cells. • Knockdown of HMGB1 inhibited AML progression under chronic stress in vivo.


Subject(s)
Gene Expression Regulation, Leukemic , HMGB1 Protein/physiology , Interleukin-1beta/physiology , Leukemia, Myeloid, Acute/physiopathology , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Neoplasm Proteins/physiology , Signal Transduction/physiology , Animals , Bone Marrow/metabolism , Cell Line, Tumor , Chronic Disease , Disease Progression , Female , Gene Knockdown Techniques , HMGB1 Protein/antagonists & inhibitors , HMGB1 Protein/biosynthesis , HMGB1 Protein/genetics , Heterografts , Humans , Inflammasomes/metabolism , Inflammation , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Leukemia, Myeloid, Acute/metabolism , Liver/metabolism , Liver/pathology , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , RNA Interference , Remission Induction , Restraint, Physical , Spleen/metabolism , Spleen/pathology , Stress, Physiological , Toll-Like Receptor 4/physiology
19.
CNS Neurosci Ther ; 27(4): 413-425, 2021 04.
Article in English | MEDLINE | ID: mdl-33034415

ABSTRACT

AIM: Spinal cord injury (SCI) is a serious disabling injury worldwide, and the excessive inflammatory response it causes plays an important role in secondary injury. Regulating the inflammatory response can be a potential therapeutic strategy for improving the prognosis of SCI. Zinc has been demonstrated to have a neuroprotective effect in experimental spinal cord injury models. In this study, we aimed to explore the neuroprotective effect of zinc through the suppression of the NLRP3 inflammasome. METHOD: Allen's method was used to establish an SCI model in C57BL/6J mice. The Basso Mouse Scale (BMS), Nissl staining were employed to confirm the protective effect of zinc on neuronal survival and functional recovery in vivo. Western blotting (WB), immunofluorescence (IF), and enzyme-linked immunosorbent assay (ELISA) were used to detect the expression levels of NLRP3 inflammasome and autophagy-related proteins. Transmission electron microscopy (TEM) was used to confirm the occurrence of zinc-induced autophagy. In vitro, lipopolysaccharide (LPS) and ATP polarized BV2 cells to a proinflammatory phenotype. 3-Methyladenine (3-MA) and bafilomycin A1 (BafA1) were chosen to explore the relationship between the NLRP3 inflammasome and autophagy. A coimmunoprecipitation assay was used to detect the ubiquitination of the NLRP3 protein. RESULTS: Our data showed that zinc significantly promoted motor function recovery after SCI. In vivo, zinc treatment inhibited the protein expression level of NLRP3 while increasing the level of autophagy. These effects were fully validated by the polarization of BV2 cells to a proinflammatory phenotype. The results showed that when 3-MA and BafA1 were applied, the promotion of autophagy by zinc was blocked and that the inhibitory effect of zinc on NLRP3 was reversed. Furthermore, co-IP confirmed that the promotion of autophagy by zinc also activated the protein expression of ubiquitin and suppressed high levels of NLRP3. CONCLUSION: Zinc provides neuroprotection by regulating NLRP3 inflammasome through autophagy and ubiquitination after SCI.


Subject(s)
Autophagy/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Neuroprotective Agents/therapeutic use , Spinal Cord Injuries/prevention & control , Ubiquitination/drug effects , Zinc/therapeutic use , Animals , Autophagy/physiology , Cell Line , Female , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , Neuroprotective Agents/pharmacology , Spinal Cord Injuries/metabolism , Ubiquitination/physiology , Zinc/pharmacology
20.
Cell Mol Neurobiol ; 41(8): 1773-1786, 2021 Nov.
Article in English | MEDLINE | ID: mdl-32968928

ABSTRACT

Parkinson's disease (PD) seriously threatens human's health. Researches have shown a close correlation between long non-coding RNAs (lncRNAs) and PD. However, the biological function of lncRNA homeobox transcript antisense RNA (HOTAIR) in PD remains largely unknown. In this study, we established PD models in vivo and in vitro by using 1-methyl-4-phenyl-2, 3, 6-tetrahydropyridine (MPTP) and 1-methyl-4-phenylpyridinium (MPP+) to assess the role of HOTAIR in pyroptotic cell death and neuronal damage. RNA immunoprecipitation (RIP) and dual luciferase reporter assay were used to verify the interaction between miR-326 and HOTAIR or ELAV like RNA binding protein 1 (ELAVL1). LncRNA HOTAIR was upregulated in PD mice and MPP+ induced SH-SY5Y cells. Additionally, knockdown of HOTAIR notably attenuated the symptom of PD in vivo. Downregulation of HOTAIR could obviously promoted cell viability and suppressed NLR family pyrin domain containing 3 (NLRP3) mediated pyroptotic cell death of SH-SY5Y cells in the presence of MPP+. Further, lncRNA HOTAIR positively regulated ELAVL1 expression by targeting miR-326, and downregulation of HOTAIR or ELAVL1 notably suppressed promotive effects of miR-326 inhibitor on MPP+ induced pyroptosis via activation of NLRP3 inflammasome. Collectively, HOTAIR silencing significantly inhibits neuronal damage through repressing NLRP3 mediated pyroptosis activation via regulation of miR-326/ELAVL1 axis in PD, which may contribute to a better understanding of PD pathogenesis and provide new treatment strategies for this disease.


Subject(s)
ELAV-Like Protein 1/biosynthesis , MicroRNAs/biosynthesis , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , Parkinsonian Disorders/metabolism , Pyroptosis/physiology , RNA, Long Noncoding/biosynthesis , 1-Methyl-4-phenylpyridinium/toxicity , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Neurons/drug effects , Neurons/metabolism , Parkinsonian Disorders/chemically induced , Pyroptosis/drug effects , RNA, Long Noncoding/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...