Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.809
Filter
2.
Biosensors (Basel) ; 14(7)2024 Jun 30.
Article in English | MEDLINE | ID: mdl-39056601

ABSTRACT

Microbial alkane degradation pathways provide biological routes for converting these hydrocarbons into higher-value products. We recently reported the functional expression of a methyl-alkylsuccinate synthase (Mas) system in Escherichia coli, allowing for the heterologous anaerobic activation of short-chain alkanes. However, the enzymatic activation of methane via natural or engineered alkylsuccinate synthases has yet to be reported. To address this, we employed high-throughput screening to engineer the itaconate (IA)-responsive regulatory protein ItcR (WT-ItcR) from Yersinia pseudotuberculosis to instead respond to methylsuccinate (MS, the product of methane addition to fumarate), resulting in genetically encoded biosensors for MS. Here, we describe ItcR variants that, when regulating fluorescent protein expression in E. coli, show increased sensitivity, improved overall response, and enhanced specificity toward exogenously added MS relative to the wild-type repressor. Structural modeling and analysis of the ItcR ligand binding pocket provide insights into the altered molecular recognition. In addition to serving as biosensors for screening alkylsuccinate synthases capable of methane activation, MS-responsive ItcR variants also establish a framework for the directed evolution of other molecular reporters, targeting longer-chain alkylsuccinate products or other succinate derivatives.


Subject(s)
Biosensing Techniques , Methane , Methane/metabolism , Escherichia coli/metabolism , Anaerobiosis , Yersinia pseudotuberculosis/metabolism , Directed Molecular Evolution , Bacterial Proteins/metabolism , Succinates/metabolism
3.
Arch Microbiol ; 206(7): 316, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38904699

ABSTRACT

Cotinine, the primary metabolite of nicotine in the human body, is an emerging pollutant in aquatic environments. It causes environmental problems and is harmful to the health of humans and other mammals; however, the mechanisms of its biodegradation have been elucidated incompletely. In this study, a novel Gram-negative strain that could degrade and utilize cotinine as a sole carbon source was isolated from municipal wastewater samples, and its cotinine degradation characteristics and kinetics were determined. Pseudomonas sp. JH-2 was able to degrade 100 mg/L (0.56 mM) of cotinine with high efficiency within 5 days at 30 ℃, pH 7.0, and 1% NaCl. Two intermediates, 6-hydroxycotinine and 6-hydroxy-3-succinoylpyridine (HSP), were identified by high-performance liquid chromatography and liquid chromatograph mass spectrometer. The draft whole genome sequence of strain JH-2 was obtained and analyzed to determine genomic structure and function. No homologs of proteins predicted in Nocardioides sp. JQ2195 and reported in nicotine degradation Pyrrolidine pathway were found in strain JH-2, suggesting new enzymes that responsible for cotinine catabolism. These findings provide meaningful insights into the biodegradation of cotinine by Gram-negative bacteria.


Subject(s)
Biodegradation, Environmental , Cotinine , Pseudomonas , Wastewater , Pseudomonas/metabolism , Pseudomonas/genetics , Pseudomonas/isolation & purification , Pseudomonas/classification , Cotinine/metabolism , Cotinine/analogs & derivatives , Wastewater/microbiology , Nicotine/metabolism , Nicotine/analogs & derivatives , Pyridines/metabolism , Genome, Bacterial , Phylogeny , Succinates
4.
Int J Mol Sci ; 25(11)2024 May 22.
Article in English | MEDLINE | ID: mdl-38891843

ABSTRACT

Mesotrione, as a widely used herbicide, is present in the environment in detectable amounts, causing serious damage. Here, we aimed to investigate the effect of mesotrione on Caco-2 cells and the possibility of its toxicity mitigation by cichoric acid. Therefore, we analyzed the cytotoxicity of both these compounds and the selected oxidative stress parameters, apoptosis and interaction of both the tested compounds with the cell membrane and their accumulation within the cells. In cytotoxicity studies, the stimulating activity of mesotrione was observed, and simultaneously, the inhibitory effect of cichoric acid was noticed. This effect was related to the results of oxidative stress analysis and apoptosis measurements. The activity level of key enzymes (glutathione peroxidase, catalase and superoxide dismutase) in Caco-2 cells exposed to cichoric acid was higher as compared to that of the control. The treatment with mesotrione did not induce apoptosis in the Caco-2 cells. The penetration of the studied compounds into the Caco-2 cells was measured by using an HPLC methodology, and the results indicate mesotrione's high penetration capacity. The distribution of charge on the surface of the cell membranes changed under the influence of both compounds. Considering the mutual interactions of beneficial and potentially toxic food ingredients, it should be noted that, despite the observed favorable trend, cichoric acid is not able to overcome the toxic and cancer-stimulating effects of this pesticide.


Subject(s)
Apoptosis , Caffeic Acids , Cyclohexanones , Oxidative Stress , Humans , Caco-2 Cells , Apoptosis/drug effects , Cyclohexanones/pharmacology , Oxidative Stress/drug effects , Caffeic Acids/pharmacology , Succinates/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adenocarcinoma/metabolism , Herbicides/toxicity , Superoxide Dismutase/metabolism , Cell Survival/drug effects , Catalase/metabolism , Glutathione Peroxidase/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism
6.
J Chem Inf Model ; 64(13): 5207-5218, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38913174

ABSTRACT

Nirmatrelvir, a pivotal component of the oral antiviral Paxlovid for COVID-19, targets the SARS-CoV-2 main protease (Mpro) as a covalent inhibitor. Here, we employed combined computational methods to explore how the prevalent Omicron variant mutation P132H, alone and in combination with A173V (P132H-A173V), affects nirmatrelvir's efficacy. Our findings suggest that P132H enhances the noncovalent binding affinity of Mpro for nirmatrelvir, whereas P132H-A173V diminishes it. Although both mutants catalyze the rate-limiting step more efficiently than the wild-type (WT) Mpro, P132H slows the overall rate of covalent bond formation, whereas P132H-A173V accelerates it. Comprehensive analysis of noncovalent and covalent contributions to the overall binding free energy of the covalent complex suggests that P132H likely enhances Mpro sensitivity to nirmatrelvir, while P132H-A173V may confer resistance. Per-residue decompositions of the binding and activation free energies pinpoint key residues that significantly affect the binding affinity and reaction rates, revealing how the mutations modulate these effects. The mutation-induced conformational perturbations alter drug-protein local contact intensities and the electrostatic preorganization of the protein, affecting noncovalent binding affinity and the stability of key reaction states, respectively. Our findings inform the mechanisms of nirmatrelvir resistance and sensitivity, facilitating improved drug design and the detection of resistant strains.


Subject(s)
Antiviral Agents , Coronavirus 3C Proteases , Mutation , SARS-CoV-2 , SARS-CoV-2/enzymology , SARS-CoV-2/drug effects , SARS-CoV-2/genetics , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/metabolism , Coronavirus 3C Proteases/chemistry , Coronavirus 3C Proteases/genetics , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Humans , COVID-19 Drug Treatment , Molecular Dynamics Simulation , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , Protease Inhibitors/metabolism , Leucine/chemistry , Thermodynamics , Sulfonamides/pharmacology , Sulfonamides/chemistry , Sulfonamides/metabolism , Protein Binding , Succinates/chemistry , Succinates/pharmacology , Succinates/metabolism , Lactams , Nitriles , Proline
7.
Int Immunopharmacol ; 137: 112531, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38906009

ABSTRACT

The role of oxidative stress and ferroptosis in osteoarthritis (OA) pathogenesis is increasingly recognized. Notably, 4-octyl Itaconate (OI) has been documented to counteract oxidative stress and inflammatory responses, highlighting its therapeutic potential in OA. This study explored the effects of OI on GPX4 methylation, oxidative stress, and ferroptosis in chondrocytes affected by OA. Our results demonstrated that OI mitigated IL-1ß-induced chondrocyte degeneration in a dose-dependent manner. It also suppressed reactive oxygen species (ROS) production and sustained GPX4 expression, thereby attenuating the degenerative impact of IL-1ß and Erastin on chondrocytes by curtailing ferroptosis. Moreover, we observed that blocking GPX4 methylation could alleviate IL-1ß-induced degeneration, oxidative stress, and ferroptosis in chondrocytes. The regulatory mechanism of OI on GPX4 expression in chondrocytes involved the inhibition of GPX4 methylation. In a mouse model of OA, OI's protective effects against OA were comparable to those of Ferrostatin-1. Thus, OI reduced chondrocyte degeneration, oxidative stress, and ferroptosis by inhibiting GPX4 methylation, offering a novel mechanistic insight into its therapeutic application in OA.


Subject(s)
Chondrocytes , Ferroptosis , Interleukin-1beta , Mice, Inbred C57BL , Osteoarthritis , Oxidative Stress , Phospholipid Hydroperoxide Glutathione Peroxidase , Succinates , Animals , Chondrocytes/drug effects , Chondrocytes/metabolism , Ferroptosis/drug effects , Oxidative Stress/drug effects , Succinates/pharmacology , Succinates/therapeutic use , Interleukin-1beta/metabolism , Osteoarthritis/drug therapy , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Mice , Male , Humans , Methylation/drug effects , Reactive Oxygen Species/metabolism , Cells, Cultured , Disease Models, Animal
8.
PLoS One ; 19(5): e0303516, 2024.
Article in English | MEDLINE | ID: mdl-38728330

ABSTRACT

Increasingly prevalent, nontuberculous mycobacteria (NTM) infections affect approximately 20% of people with cystic fibrosis (CF). Previous studies of CF sputum identified lower levels of the host metabolite itaconate in those infected with NTM. Itaconate can inhibit the growth of M. tuberculosis (MTB) in vitro via the inhibition of the glyoxylate cycle enzyme (ICL), but its impact on NTM is unclear. To test itaconic acid's (IA) effect on NTM growth, laboratory and CF clinical strains of Mycobacterium abscessus and Mycobacterium avium were cultured in 7H9 minimal media supplemented with 1-10 mM of IA and short-chain fatty acids (SCFA). M. avium and M. abscessus grew when supplemented with SCFAs, whereas the addition of IA (≥ 10 mM) completely inhibited NTM growth. NTM supplemented with acetate or propionate and 5 mM IA displayed slower growth than NTM cultured with SCFA and ≤ 1 mM of IA. However, IA's inhibition of NTM was pH dependent; as similar and higher quantities (100 mM) of pH adjusted IA (pH 7) did not inhibit growth in vitro, while in an acidic minimal media (pH 6.1), 1 to 5 mM of non-pH adjusted IA inhibited growth. None of the examined isolates displayed the ability to utilize IA as a carbon source, and IA added to M. abscessus isocitrate lyase (ICL) decreased enzymatic activity. Lastly, the addition of cell-permeable 4-octyl itaconate (4-OI) to THP-1 cells enhanced NTM clearance, demonstrating a potential role for IA/itaconate in host defense against NTM infections.


Subject(s)
Succinates , Succinates/pharmacology , Succinates/metabolism , Humans , Hydrogen-Ion Concentration , Nontuberculous Mycobacteria/drug effects , Nontuberculous Mycobacteria/growth & development , THP-1 Cells , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium avium/drug effects , Mycobacterium avium/growth & development , Mycobacterium abscessus/drug effects , Mycobacterium abscessus/growth & development , Mycobacterium abscessus/metabolism
9.
Clin Immunol ; 264: 110255, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38763433

ABSTRACT

Fibroblast-like synoviocytes (FLS) play critical roles in rheumatoid arthritis (RA). Itaconate (ITA), an endogenous metabolite derived from the tricarboxylic acid (TCA) cycle, has attracted attention because of its anti-inflammatory, antiviral, and antimicrobial effects. This study evaluated the effect of ITA on FLS and its potential to treat RA. ITA significantly decreased FLS proliferation and migration in vitro, as well as mitochondrial oxidative phosphorylation and glycolysis measured by an extracellular flux analyzer. ITA accumulates metabolites including succinate and citrate in the TCA cycle. In rats with type II collagen-induced arthritis (CIA), intra-articular injection of ITA reduced arthritis and bone erosion. Irg1-deficient mice lacking the ability to produce ITA had more severe arthritis than control mice in the collagen antibody-induced arthritis. ITA ameliorated CIA by inhibiting FLS proliferation and migration. Thus, ITA may be a novel therapeutic agent for RA.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Cell Movement , Cell Proliferation , Fibroblasts , Succinates , Synoviocytes , Animals , Synoviocytes/drug effects , Synoviocytes/metabolism , Cell Movement/drug effects , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Cell Proliferation/drug effects , Succinates/pharmacology , Rats , Fibroblasts/drug effects , Fibroblasts/metabolism , Male , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Mice , Mice, Knockout , Cells, Cultured , Mice, Inbred DBA , Citric Acid Cycle/drug effects
10.
Eur J Pharmacol ; 976: 176696, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38821160

ABSTRACT

Cichoric acid (CA), a widely utilized polyphenolic compound in medicine, has garnered significant attention due to its potential health benefits. Sepsis-induced acute kidney disease (AKI) is related with an elevated risk of end-stage kidney disease (ESKD). However, it remains unclear whether CA provides protection against septic AKI. The aim of this study is to investigated the protective effect and possible mechanisms of CA against LPS-induced septic AKI. Sepsis-induced AKI was induced in mice through intraperitoneal injection of lipopolysaccharide (LPS), and RAW264.7 macrophages were incubated with LPS. LPS exposure significantly increased the levels of M1 macrophage biomarkers while reducing the levels of M2 macrophage indicators. This was accompanied by the release of inflammatory factors, superoxide anion production, mitochondrial dysfunction, activation of succinate dehydrogenase (SDH), and subsequent succinate formation. Conversely, pretreatment with CA mitigated these abnormalities. CA attenuated hypoxia-inducible factor-1α (HIF-1α)-induced glycolysis by lifting the NAD+/NADH ratio in macrophages. Additionally, CA disrupted the K (lysine) acetyltransferase 2A (KAT2A)/α-tubulin complex, thereby reducing α-tubulin acetylation and subsequently inactivating the NLRP3 inflammasome. Importantly, administration of CA ameliorated LPS-induced renal pathological damage, apoptosis, inflammation, oxidative stress, and disturbances in mitochondrial function in mice. Overall, CA restrained HIF-1α-mediated glycolysis via inactivation of SDH, leading to NLRP3 inflammasome inactivation and the amelioration of sepsis-induced AKI.


Subject(s)
Acute Kidney Injury , Caffeic Acids , Lipopolysaccharides , Macrophages , NLR Family, Pyrin Domain-Containing 3 Protein , Sepsis , Succinates , Animals , Sepsis/complications , Sepsis/drug therapy , Mice , Acute Kidney Injury/drug therapy , Acute Kidney Injury/metabolism , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Male , Succinates/pharmacology , Succinates/therapeutic use , Macrophages/drug effects , Macrophages/metabolism , Caffeic Acids/pharmacology , Caffeic Acids/therapeutic use , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , RAW 264.7 Cells , Oxidative Stress/drug effects , Inflammasomes/metabolism , Mice, Inbred C57BL , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Glycolysis/drug effects , Apoptosis/drug effects , Kidney/pathology , Kidney/drug effects , Kidney/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Macrophage Activation/drug effects
11.
Int Immunopharmacol ; 135: 112277, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38788445

ABSTRACT

Sepsis, a systemic inflammatory response triggered by infection, has a considerably high mortality rate. However, effective prevention and intervention measures against sepsis remain insufficient. Therefore, this study aimed to investigate the mechanisms underlying the protective properties of immune response gene-1 (IRG1) and 4-Octyl itaconate (OI) during acute liver damage in mice with sepsis. A sepsis mouse model was established to compare wild-type and IRG1-/- groups. The impact of IRG1/Itaconate on pro- and anti-inflammatory cytokines was evaluated using J774A.1 cells. IRG1/Itaconate substantially reduced pro-inflammatory cytokines and increased the release of anti-inflammatory cytokines. It reduced pathological damage to liver tissues, preserved normal liver function, decreased the release of reactive oxygen species (ROS) and LDH, and enhanced the GSH/GSSG ratio. Moreover, IRG1 and itaconic acid activated the Nrf2 signaling pathway, regulating the expression of its downstream antioxidative stress-related proteins. Additionally, they inhibited the activity of NLRP3 inflammatory vesicles to suppress the expression of macrophage-associated pyroptosis signaling molecules. Our findings demonstrate that IRG1/OI inhibits NLRP3 inflammatory vesicle activation and macrophage pyroptosis by modulating the Nrf2 signaling pathway, thereby attenuating acute liver injury in mice with sepsis. These findings could facilitate the clinical application of IRG1/Itaconate to prevent sepsis-induced acute liver injury.


Subject(s)
Macrophages , Mice, Inbred C57BL , NF-E2-Related Factor 2 , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Sepsis , Signal Transduction , Succinates , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Succinates/therapeutic use , Succinates/pharmacology , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Sepsis/drug therapy , Sepsis/complications , Sepsis/immunology , Pyroptosis/drug effects , Mice , Signal Transduction/drug effects , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Knockout , Liver/pathology , Liver/drug effects , Liver/metabolism , Liver/immunology , Cell Line , Disease Models, Animal , Cytokines/metabolism , Hydro-Lyases/metabolism , Reactive Oxygen Species/metabolism , Humans , Carboxy-Lyases/metabolism , Carboxy-Lyases/genetics
12.
Biotechnol Lett ; 46(4): 593-600, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38809464

ABSTRACT

Itaconic acid is an excellent polymeric precursor with a wide range of industrial applications. The efficient production of itaconate from various renewable substrates was demonstrated by engineered Escherichia coli. However, limitation in the itaconic acid precursor supply was revealed by finding out the key intermediate of the tricarboxylic acid in the itaconic acid pathway. Efforts of enhancing the cis-aconitate flux and preserving the isocitrate pool to increase itaconic acid productivity are required. In this study, we introduce a synthetic protein scaffold system between CadA and AcnA to physically combine the two enzymes. Through the introduction of a synthetic protein scaffold, 2.1 g L-1 of itaconic acid was produced at pH 7 and 37 °C. By fermentation, 20.1 g L-1 for 48 h of itaconic acid was produced with a yield of 0.34 g g-1 glycerol. These results suggest that carbon flux was successfully increased itaconic acid productivity.


Subject(s)
Escherichia coli , Metabolic Engineering , Succinates , Succinates/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Metabolic Engineering/methods , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Fermentation
13.
Cell Rep ; 43(5): 114142, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38691458

ABSTRACT

Despite medical advances, there remains an unmet need for better treatment of obesity. Itaconate, a product of the decarboxylation of the tricarboxylic acid cycle intermediate cis-aconitate, plays a regulatory role in both metabolism and immunity. Here, we show that itaconate, as an endogenous compound, counteracts high-fat-diet (HFD)-induced obesity through leptin-independent mechanisms in three mouse models. Specifically, itaconate reduces weight gain, reverses hyperlipidemia, and improves glucose tolerance in HFD-fed mice. Additionally, itaconate enhances energy expenditure and the thermogenic capacity of brown adipose tissue (BAT). Unbiased proteomic analysis reveals that itaconate upregulates key proteins involved in fatty acid oxidation and represses the expression of lipogenic genes. Itaconate may provoke a major metabolic reprogramming by inducing fatty acid oxidation and suppression of fatty acid synthesis in BAT. These findings highlight itaconate as a potential activator of BAT-mediated thermogenesis and a promising candidate for anti-obesity therapy.


Subject(s)
Adipocytes, Brown , Diet, High-Fat , Mice, Inbred C57BL , Obesity , Succinates , Thermogenesis , Animals , Thermogenesis/drug effects , Obesity/metabolism , Obesity/drug therapy , Succinates/pharmacology , Diet, High-Fat/adverse effects , Mice , Male , Adipocytes, Brown/metabolism , Adipocytes, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/drug effects , Energy Metabolism/drug effects
14.
Nat Commun ; 15(1): 4096, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750019

ABSTRACT

The presence of heterogeneity in responses to oncolytic virotherapy poses a barrier to clinical effectiveness, as resistance to this treatment can occur through the inhibition of viral spread within the tumor, potentially leading to treatment failures. Here we show that 4-octyl itaconate (4-OI), a chemical derivative of the Krebs cycle-derived metabolite itaconate, enhances oncolytic virotherapy with VSVΔ51 in various models including human and murine resistant cancer cell lines, three-dimensional (3D) patient-derived colon tumoroids and organotypic brain tumor slices. Furthermore, 4-OI in combination with VSVΔ51 improves therapeutic outcomes in a resistant murine colon tumor model. Mechanistically, we find that 4-OI suppresses antiviral immunity in cancer cells through the modification of cysteine residues in MAVS and IKKß independently of the NRF2/KEAP1 axis. We propose that the combination of a metabolite-derived drug with an oncolytic virus agent can greatly improve anticancer therapeutic outcomes by direct interference with the type I IFN and NF-κB-mediated antiviral responses.


Subject(s)
Oncolytic Virotherapy , Oncolytic Viruses , Succinates , Animals , Humans , Oncolytic Virotherapy/methods , Succinates/pharmacology , Mice , Cell Line, Tumor , Interferon Type I/metabolism , NF-E2-Related Factor 2/metabolism , Colonic Neoplasms/therapy , Colonic Neoplasms/immunology , Colonic Neoplasms/drug therapy , Antiviral Agents/pharmacology , NF-kappa B/metabolism , I-kappa B Kinase/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Inflammation/drug therapy , Female , Vesicular stomatitis Indiana virus/physiology , Vesicular stomatitis Indiana virus/drug effects , Signal Transduction/drug effects
15.
Ann Clin Transl Neurol ; 11(7): 1732-1749, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38738556

ABSTRACT

OBJECTIVE: Neuromyelitis optica spectrum disorders (NMOSD) are rare inflammatory astrocytic diseases of the central nervous system (CNS). The roles of immune response gene-1 (IRG1) and the IRG1-itaconic acid-NLRP3 inflammatory pathway in the pathogenesis of NMOSD and the effects of 4-octyl itaconate (4-OI) on the NLRP3 inflammatory pathway in NMOSD are unclear. This study aimed to determine the role of IRG1 and the activation status of the NLRP3 inflammatory pathway in acute-onset NMOSD and to investigate the inhibitory effects of 4-OI on NLRP3 inflammasome activation via the IRG1-itaconic acid-NLRP3 pathway in monocytes and macrophages by using in vitro models. METHODS: Peripheral blood mononuclear cells (PBMCs) and serum were collected from patients with acute NMOSDs and healthy controls (HC), followed by monocyte typing and detection of the expression of NLRP3-related inflammatory factors. Subsequently, the effects of 4-OI on the IRG1-itaconic acid-NLRP3 pathway were investigated in peripheral monocytes from patients with NMOSD and in macrophages induced by human myeloid leukemia mononuclear cells (THP-1 cells) via in vitro experiments. RESULTS: Patients with acute NMOSD exhibited upregulated IRG1 expression. In particular, the upregulation of the expression of the NLRP3 inflammasome and proinflammatory factors was notable in monocytes in acute NMOSD patients. 4-OI inhibited the activation of the IRG1-itaconic acid-NLRP3 inflammatory pathway in the PBMCs of patients with NMOSD. INTERPRETATION: 4-OI could effectively inhibit NLRP3 signaling, leading to the inhibition of proinflammatory cytokine production in patients with NMOSD-derived PBMCs and in a human macrophage model. Thus, 4-OI and itaconate could have important therapeutic value for the treatment of NMOSD in the future.


Subject(s)
NLR Family, Pyrin Domain-Containing 3 Protein , Neuromyelitis Optica , Succinates , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Neuromyelitis Optica/drug therapy , Succinates/pharmacology , Female , Male , Adult , Middle Aged , Inflammasomes/drug effects , Inflammasomes/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Inflammation/drug therapy , Signal Transduction/drug effects , Macrophages/drug effects , Macrophages/metabolism , Carboxy-Lyases
16.
Cell Mol Immunol ; 21(7): 689-706, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38745069

ABSTRACT

Cytokine storm syndrome (CSS) is a life-threatening systemic inflammatory syndrome involving innate immune hyperactivity triggered by various therapies, infections, and autoimmune conditions. However, the potential interplay between innate immune cells is not fully understood. Here, using poly I:C and lipopolysaccharide (LPS)-induced cytokine storm models, a protective role of neutrophils through the modulation of macrophage activation was identified in a CSS model. Intravital imaging revealed neutrophil-derived extracellular vesicles (NDEVs) in the liver and spleen, which were captured by macrophages. NDEVs suppressed proinflammatory cytokine production by macrophages when cocultured in vitro or infused into CSS models. Metabolic profiling of macrophages treated with NDEV revealed elevated levels of the anti-inflammatory metabolite, itaconate, which is produced from cis-aconitate in the Krebs cycle by cis-aconitate decarboxylase (Acod1, encoded by Irg1). Irg1 in macrophages, but not in neutrophils, was critical for the NDEV-mediated anti-inflammatory effects. Mechanistically, NDEVs delivered miR-27a-3p, which suppressed the expression of Suclg1, the gene encoding the enzyme that metabolizes itaconate, thereby resulting in the accumulation of itaconate in macrophages. These findings demonstrated that neutrophil-to-macrophage communication mediated by extracellular vesicles is critical for promoting the anti-inflammatory reprogramming of macrophages in CSS and may have potential implications for the treatment of this fatal condition.


Subject(s)
Cytokine Release Syndrome , Extracellular Vesicles , Macrophages , Neutrophils , Succinates , Animals , Extracellular Vesicles/metabolism , Succinates/metabolism , Macrophages/metabolism , Macrophages/immunology , Neutrophils/metabolism , Neutrophils/immunology , Mice , Cytokine Release Syndrome/metabolism , Carboxy-Lyases/metabolism , Mice, Inbred C57BL , Cell Communication , MicroRNAs/metabolism , MicroRNAs/genetics , Cytokines/metabolism , Male , Disease Models, Animal , Hydro-Lyases
17.
Biomed Pharmacother ; 176: 116814, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38820974

ABSTRACT

Diabetes and derived complications, especially diabetic nephropathy and neuropathy annually cause great morbimortality worldwide. 5-hydroxytryptamine (5-HT) acts as a modulator of renal sympathetic input and vascular tone. In this line, 5-HT2 receptor blockade has been linked with reduced incidence and progression of diabetic microvascular alterations. In this work, we aimed to determine, in diabetic rats, whether 5-HT2 blockade ameliorates renal function and to characterize the serotonergic modulatory action on renal sympathetic neurotransmission. Diabetes was induced in male Wistar rats by alloxan administration (150 mg/kg, s.c.), and sarpogrelate (30 mg/kg·day, p.o.; 5-HT2 antagonist) was administered for 14 days (DM-S). Normoglycemic and diabetic (DM) animals were maintained as aged-matched controls. At 28th day, DM-S animals were anesthetized and prepared for the in situ autoperfusion of the kidney. Renal vasoconstrictor responses were induced electrically or by i.a. noradrenaline (NA) administration. The role of 5-HT and selective 5-HT agonist/antagonist were studied on these renal vasopressor responses. Sarpogrelate treatment decreased renal sympathetic-induced vasopressor responses, reduced renal hypertrophy and kidney damage markers increased in DM. Intraarterial 5-HT inhibited the sympathetic-induced renal vasoconstrictions, effect reproduced by 5-CT, AS-19, L-694,247 and LY 344864 (5-HT1/5/7, 5-HT7, 5-HT1D and 5-HT1F receptor agonists, respectively). Blocking 5-HT1D/1F/7 receptors completely abolished the 5-CT sympatho-inhibition. NA vasoconstrictions were not altered by any of the 5-HT agonists tested. Thus, in experimental diabetes, chronic sarpogrelate treatment reduces renal damage markers, kidney hypertrophy and renal sympathetic hyperactivity and modifies serotonergic modulation of renal sympathetic neurotransmission, causing a sympatho-inhibition by prejunctional 5-HT1D/1F and 5-HT7 activation.


Subject(s)
Diabetes Mellitus, Experimental , Kidney , Rats, Wistar , Succinates , Sympathetic Nervous System , Animals , Succinates/pharmacology , Male , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/physiopathology , Kidney/drug effects , Kidney/innervation , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiopathology , Rats , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Serotonin/metabolism , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/physiopathology , Vasoconstriction/drug effects
18.
J Environ Manage ; 360: 121100, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38744205

ABSTRACT

Removal of heavy metals using the electrokinetic (EK) remediation technology is restricted by soils containing a fraction of clay particles above 12%. Furthermore, it is also affected by hydroxide precipitation (focusing phenomenon) close to the cathode. A modified EK reactor containing a permeable reactive barrier (PRB) was proposed herein where the enzyme-induced carbonate precipitation (EICP) treatment was incorporated into the PRB. Despite that, NH4+-N pollution induced by the urea hydrolysis resulting from the EICP treatment causes serious threats to surrounding environments and human health. There were four types of tests applied to the present work, including CP, TS1, TS2, and TS3 tests. CP test neglected the bio-PRB, while TS1 test considered the bio-PRB. TS2 test based on TS1 test tackled NH4+-N pollution using the struvite precipitation technology. TS3 test based on TS2 test applied EDDS to enhance the removal of Cu and Pb. In CP test, the removal efficiency applied to Cu and Pb removals was as low as approximately 10%, presumably due to the focusing phenomenon. The removal efficiency was elevated to approximately 24% when the bio-PRB and the electrolyte reservoir were involved in TS1 test. TS2 test indicated that the rate of struvite precipitation was 40 times faster than the ureolysis rate, meaning that the struvite precipitate had sequestered NH4+ before it started threatening surrounding environments. The chelation between Cu2+ and EDDS took place when EDDS played a part in TS3 test. It made Cu2+ negatively surface charged by transforming Cu2+ into EDDSCu2-. The chelation caused those left in S4 and S4 to migrate toward the bio-PRB, whereas it also caused those left in S1 and S2 to migrate toward the anode. Due to this reason, the fraction of Cu2+ removed by the bio-PRB and the electrolyte reservoir is raised to 32% and 26% respectively, and the fraction of remaining Cu was reduced to 41%. Also, the removal efficiency applied to Pb removal was raised to 50%. Results demonstrate the potential of struvite and EDDS-assisted EK-PRB technology as a cleanup method for Cu- and Pb-contaminated loess.


Subject(s)
Copper , Lead , Struvite , Copper/chemistry , Lead/chemistry , Struvite/chemistry , Soil/chemistry , Succinates/chemistry , Soil Pollutants/chemistry
19.
J Neuroinflammation ; 21(1): 132, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760862

ABSTRACT

BACKGROUND: Neonatal hypoxic-ischemic encephalopathy (HIE) is one of the most common neurological problems occurring in the perinatal period. However, there still is not a promising approach to reduce long-term neurodevelopmental outcomes of HIE. Recently, itaconate has been found to exhibit anti-oxidative and anti-inflammatory effects. However, the therapeutic efficacy of itaconate in HIE remains inconclusive. Therefore, this study attempts to explore the pathophysiological mechanisms of oxidative stress and inflammatory responses in HIE as well as the potential therapeutic role of a derivative of itaconate, 4-octyl itaconate (4OI). METHODS: We used 7-day-old mice to induce hypoxic-ischemic (HI) model by right common carotid artery ligation followed by 1 h of hypoxia. Behavioral experiments including the Y-maze and novel object recognition test were performed on HI mice at P60 to evaluate long-term neurodevelopmental outcomes. We employed an approach combining non-targeted metabolomics with transcriptomics to screen alterations in metabolic profiles and gene expression in the hippocampal tissue of the mice at 8 h after hypoxia. Immunofluorescence staining and RT-PCR were used to evaluate the pathological changes in brain tissue cells and the expression of mRNA and proteins. 4OI was intraperitoneally injected into HI model mice to assess its anti-inflammatory and antioxidant effects. BV2 and C8D1A cells were cultured in vitro to study the effect of 4OI on the expression and nuclear translocation of Nrf2. We also used Nrf2-siRNA to further validate 4OI-induced Nrf2 pathway in astrocytes. RESULTS: We found that in the acute phase of HI, there was an accumulation of pyruvate and lactate in the hippocampal tissue, accompanied by oxidative stress and pro-inflammatory, as well as increased expression of antioxidative stress and anti-inflammatory genes. Treatment of 4OI could inhibit activation and proliferation of microglial cells and astrocytes, reduce neuronal death and relieve cognitive dysfunction in HI mice. Furthermore, 4OI enhanced nuclear factor erythroid-2-related factor (Nfe2l2; Nrf2) expression and nuclear translocation in astrocytes, reduced pro-inflammatory cytokine production, and increased antioxidant enzyme expression. CONCLUSION: Our study demonstrates that 4OI has a potential therapeutic effect on neuronal damage and cognitive deficits in HIE, potentially through the modulation of inflammation and oxidative stress pathways by Nrf2 in astrocytes.


Subject(s)
Animals, Newborn , Astrocytes , Hypoxia-Ischemia, Brain , NF-E2-Related Factor 2 , Neuroprotective Agents , Succinates , Animals , NF-E2-Related Factor 2/metabolism , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/pathology , Mice , Astrocytes/drug effects , Astrocytes/metabolism , Succinates/pharmacology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Signal Transduction/drug effects , Mice, Inbred C57BL , Oxidative Stress/drug effects , Oxidative Stress/physiology , Disease Models, Animal
20.
Respir Res ; 25(1): 205, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730297

ABSTRACT

BACKGROUND: Obesity is the main risk factor leading to the development of various respiratory diseases, such as asthma and pulmonary hypertension. Pulmonary microvascular endothelial cells (PMVECs) play a significant role in the development of lung diseases. Aconitate decarboxylase 1 (Acod1) mediates the production of itaconate, and Acod1/itaconate axis has been reported to play a protective role in multiple diseases. However, the roles of Acod1/itaconate axis in the PMVECs of obese mice are still unclear. METHODS: mRNA-seq was performed to identify the differentially expressed genes (DEGs) between high-fat diet (HFD)-induced PMVECs and chow-fed PMVECs in mice (|log2 fold change| ≥ 1, p ≤ 0.05). Free fatty acid (FFA) was used to induce cell injury, inflammation and mitochondrial oxidative stress in mouse PMVECs after transfection with the Acod1 overexpressed plasmid or 4-Octyl Itaconate (4-OI) administration. In addition, we investigated whether the nuclear factor erythroid 2-like 2 (Nrf2) pathway was involved in the effects of Acod1/itaconate in FFA-induced PMVECs. RESULTS: Down-regulated Acod1 was identified in HFD mouse PMVECs by mRNA-seq. Acod1 expression was also reduced in FFA-treated PMVECs. Acod1 overexpression inhibited cell injury, inflammation and mitochondrial oxidative stress induced by FFA in mouse PMVECs. 4-OI administration showed the consistent results in FFA-treated mouse PMVECs. Moreover, silencing Nrf2 reversed the effects of Acod1 overexpression and 4-OI administration in FFA-treated PMVECs, indicating that Nrf2 activation was required for the protective effects of Acod1/itaconate. CONCLUSION: Our results demonstrated that Acod1/Itaconate axis might protect mouse PMVECs from FFA-induced injury, inflammation and mitochondrial oxidative stress via activating Nrf2 pathway. It was meaningful for the treatment of obesity-caused pulmonary microvascular endotheliopathy.


Subject(s)
Carboxy-Lyases , Endothelial Cells , Lung , Mice, Inbred C57BL , NF-E2-Related Factor 2 , Obesity , Succinates , Animals , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Mice , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Endothelial Cells/pathology , Carboxy-Lyases/metabolism , Carboxy-Lyases/genetics , Obesity/metabolism , Obesity/complications , Male , Succinates/pharmacology , Lung/metabolism , Lung/drug effects , Lung/pathology , Lung/blood supply , Cells, Cultured , Microvessels/metabolism , Microvessels/drug effects , Microvessels/pathology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Diet, High-Fat/adverse effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Hydro-Lyases
SELECTION OF CITATIONS
SEARCH DETAIL