Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.088
Filter
1.
Bull Exp Biol Med ; 177(2): 181-184, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39090467

ABSTRACT

We performed a comprehensive study of protein (total protein, medium-molecular-weight peptides, creatinine, and urea), purine (uric acid), and lipid (cholesterol, triglycerides) metabolism, activity of AST, ALT, and acid phosphatase in blood plasma of white male rats under conditions of restriction of motor activity up to 28 days. Patterns of changes in metabolic profile during hypokinesia were established: prevalence of catabolic processes and atherogenic shifts in the lipid spectrum with maximum manifestation on 14-21 days of the experiment.


Subject(s)
Cholesterol , Triglycerides , Animals , Male , Rats , Triglycerides/blood , Triglycerides/metabolism , Cholesterol/blood , Cholesterol/metabolism , Uric Acid/blood , Uric Acid/metabolism , Motor Activity/physiology , Metabolome/physiology , Lipid Metabolism/physiology , Aspartate Aminotransferases/blood , Aspartate Aminotransferases/metabolism , Alanine Transaminase/blood , Alanine Transaminase/metabolism , Creatinine/blood , Acid Phosphatase/metabolism , Acid Phosphatase/blood , Urea/blood , Hypokinesia/metabolism , Hypokinesia/physiopathology
2.
Sci Rep ; 14(1): 18146, 2024 08 05.
Article in English | MEDLINE | ID: mdl-39103417

ABSTRACT

Urate nephropathy, a common complication of hyperuricemia, has garnered increasing attention worldwide. However, the exact pathogenesis of this condition remains unclear. Currently, inflammation is widely accepted as the key factor in urate nephropathy. Therefore, the aim of this study was to elucidate the interaction of lincRNA-p21/AIF-1/CMPK2/NLRP3 via exosomes in urate nephropathy. This study evaluated the effect of lincRNA-p21/AIF-1/CMPK2/NLRP3 using clinical data collected from patients with urate nephropathy and human renal tubular epithelial cells (HK2) cultured with different concentrations of urate. In clinical research section, the level of lincRNA-p21/AIF-1 in exosomes of urine in patients with hyperuricemia or urate nephropathy was found to be increased, particularly in patients with urate nephropathy. In vitro study section, the level of exosomes, inflammation, autophagy, and apoptosis was increased in HK2 cells induced by urate. Additionally, the expression of lincRNA-p21, AIF-1, CMPK2, and NLRP3 was upregulated in exosomes and HK2 cells. Furthermore, manipulating the activity of lincRNA-p21, AIF-1, CMPK2, and NLRP3 through overexpression or interference vectors regulated the level of inflammation, autophagy, and apoptosis in HK2 cells. In conclusion, the pathway of lincRNA-p21/AIF-1/CMPK2/NLRP3 contributed to inflammation, autophagy, and apoptosis of human renal tubular epithelial cell induced by urate via exosomes. Additionally, the specific exosomes in urine might serve as novel biomarkers for urate nephropathy.


Subject(s)
Apoptosis , Autophagy , Epithelial Cells , Exosomes , NLR Family, Pyrin Domain-Containing 3 Protein , RNA, Long Noncoding , Uric Acid , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Uric Acid/metabolism , Exosomes/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , RNA, Long Noncoding/metabolism , RNA, Long Noncoding/genetics , Signal Transduction , Inflammation/metabolism , Inflammation/pathology , Kidney Tubules/metabolism , Kidney Tubules/pathology , Cell Line , Male , Apoptosis Inducing Factor/metabolism , Female , Middle Aged , Hyperuricemia/metabolism , Hyperuricemia/urine , Calcium-Binding Proteins , Microfilament Proteins
3.
Biomolecules ; 14(7)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39062467

ABSTRACT

Hyperuricemia (HUA) may lead to myocardial cell damage, thereby promoting the occurrence and adverse outcomes of heart diseases. In this review, we discuss the latest clinical research progress, and explore the impact of HUA on myocardial damage-related diseases such as myocardial infarction, arrhythmias, and heart failure. We also combined recent findings from basic research to analyze potential mechanisms linking HUA with myocardial injury. In different pathological models (such as direct action of high uric acid on myocardial cells or combined with myocardial ischemia-reperfusion model), HUA may cause damage by activating the NOD-like receptor protein 3 inflammasome-induced inflammatory response, interfering with cardiac cell energy metabolism, affecting antioxidant defense systems, and stimulating reactive oxygen species production to enhance the oxidative stress response, ultimately resulting in decreased cardiac function. Additionally, we discuss the impact of lowering uric acid intervention therapy and potential safety issues that may arise. However, as the mechanism underlying HUA-induced myocardial injury is poorly defined, further research is warranted to aid in the development novel therapeutic strategies for HUA-related cardiovascular diseases.


Subject(s)
Heart Diseases , Hyperuricemia , Oxidative Stress , Uric Acid , Humans , Hyperuricemia/metabolism , Hyperuricemia/drug therapy , Hyperuricemia/complications , Animals , Heart Diseases/metabolism , Uric Acid/metabolism , Reactive Oxygen Species/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism
4.
PLoS One ; 19(7): e0307181, 2024.
Article in English | MEDLINE | ID: mdl-39046973

ABSTRACT

Metabolic-associated fatty liver disease (MAFLD) is predominantly associated with metabolic disturbances representing aberrant liver function and increased uric acid (UA) levels. Growing evidences have suggested a close relationship between metabolic disturbances and the gut microbiota. A placebo-controlled, double-blinded, randomized clinical trial was therefore conducted to explore the impacts of daily supplements with various combinations of the probiotics, Lactobacillus fermentum TSF331, Lactobacillus reuteri TSR332, and Lactobacillus plantarum TSP05 with a focus on liver function and serum UA levels. Test subjects with abnormal levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT), and UA were recruited and randomly allocated into six groups. Eighty-two participants successfully completed the 60-day intervention without any dropouts or occurrence of adverse events. The serum AST, ALT, and UA levels were significantly reduced in all treatment groups (P < 0.05). The fecal microbiota analysis revealed the intervention led to an increase in the population of commensal bacteria and a decrease in pathobiont bacteria, especially Bilophila wadsworthia. The in vitro study indicated the probiotic treatments reduced lipid accumulation and inflammatory factor expressions in HepG2 cells, and also promoted UA excretion in Caco-2 cells. The supplementation of multi-strain probiotics (TSF331, TSR332, and TSP05) together can improve liver function and UA management and may have good potential in treating asymptomatic MAFLD. Trial registration. The trial was registered in the US Library of Medicine (clinicaltrials.gov) with the number NCT06183801 on December 28, 2023.


Subject(s)
Lactobacillus plantarum , Limosilactobacillus fermentum , Limosilactobacillus reuteri , Probiotics , Uric Acid , Humans , Probiotics/administration & dosage , Lactobacillus plantarum/physiology , Male , Uric Acid/blood , Uric Acid/metabolism , Female , Pilot Projects , Middle Aged , Double-Blind Method , Liver/metabolism , Adult , Gastrointestinal Microbiome/drug effects , Hep G2 Cells , Caco-2 Cells , Aspartate Aminotransferases/blood , Feces/microbiology , Alanine Transaminase/blood
5.
Nat Rev Rheumatol ; 20(8): 510-523, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38992217

ABSTRACT

The pathogenesis of gout involves a series of steps beginning with hyperuricaemia, followed by the deposition of monosodium urate crystal in articular structures and culminating in an innate immune response, mediated by the NLRP3 inflammasome, to the deposited crystals. Large genome-wide association studies (GWAS) of serum urate levels initially identified the genetic variants with the strongest effects, mapping mainly to genes that encode urate transporters in the kidney and gut. Other GWAS highlighted the importance of uncommon genetic variants. More recently, genetic and epigenetic genome-wide studies have revealed new pathways in the inflammatory process of gout, including genetic associations with epigenomic modifiers. Epigenome-wide association studies are also implicating epigenomic remodelling in gout, which perhaps regulates the responsiveness of the innate immune system to monosodium urate crystals. Notably, genes implicated in gout GWAS do not include those encoding components of the NLRP3 inflammasome itself, but instead include genes encoding molecules involved in its regulation. Knowledge of the molecular mechanisms underlying gout has advanced through the translation of genetic associations into specific molecular mechanisms. Notable examples include ABCG2, HNF4A, PDZK1, MAF and IL37. Current genetic studies are dominated by participants of European ancestry; however, studies focusing on other population groups are discovering informative population-specific variants associated with gout.


Subject(s)
Genome-Wide Association Study , Gout , Gout/genetics , Humans , Epigenomics/methods , Genetic Predisposition to Disease , Epigenesis, Genetic , Transcriptome , Uric Acid/blood , Uric Acid/metabolism , Hyperuricemia/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics
6.
Neuropsychopharmacol Hung ; 26(2): 105-124, 2024 06.
Article in English | MEDLINE | ID: mdl-38994859

ABSTRACT

Major mood disorder (i.e. major depressive disorder [MDD] and bipolar disorders [BPDs]) are among the most prevalent and disabling mental illnesses. Several, frequently intertwining theories (such as the monoamine, neuroinflammatory and neurotrophic theories) exist to explain the etiopathogenic background of mood disorders. A lesser-known hypothesis addresses the role of oxidative stress (OS; i.e. the overproduction and accumulation of free radicals) in the pathogenesis of these mental disorders. Free radicals are capable of damaging phospholipids, polyunsaturated fatty acids, proteins and nucleic acids. In the brain, OS impairs inter alia synaptic signalling and neuroplasticity. In the current paper, in addition to a brief description of the aforementioned pathophysiological processes involved in mood disorders (with a special focus on OS), we discuss in detail the results of studies on changes in non-enzymatic antioxidant uric acid (UA) levels in major mood disorders. Findings to date indicate that UA - a routinely measured laboratory parameter - may be a candidate biomarker to distinguish between MDD and BPD. Since the diagnostic criteria are identical for major depressive episodes regardless of whether the episode occurs in the context of MDD or BPD and also bearing in mind that the treatment for those two disorders is different, we may conclude that the identification of biomarkers to enable MDD to be distinguished from BPD would be of great clinical relevance.


Subject(s)
Bipolar Disorder , Depressive Disorder, Major , Oxidative Stress , Uric Acid , Humans , Uric Acid/metabolism , Depressive Disorder, Major/metabolism , Bipolar Disorder/metabolism , Mood Disorders/metabolism , Biomarkers/metabolism , Brain/metabolism
7.
Commun Biol ; 7(1): 828, 2024 Jul 07.
Article in English | MEDLINE | ID: mdl-38972919

ABSTRACT

Crystallization of monosodium urate monohydrate (MSU) leads to painful gouty arthritis. Despite extensive research it is still unknown how this pathological biomineralization occurs, which hampers its prevention. Here we show how inflammatory MSU crystals form after a non-inflammatory amorphous precursor (AMSU) that nucleates heterogeneously on collagen fibrils from damaged articular cartilage of gout patients. This non-classical crystallization route imprints a nanogranular structure to biogenic acicular MSU crystals, which have smaller unit cell volume, lower microstrain, and higher crystallinity than synthetic MSU. These distinctive biosignatures are consistent with the template-promoted crystallization of biotic MSU crystals after AMSU at low supersaturation, and their slow growth over long periods of time (possibly years) in hyperuricemic gout patients. Our results help to better understand gout pathophysiology, underline the role of cartilage damage in promoting MSU crystallization, and suggest that there is a time-window to treat potential gouty patients before a critical amount of MSU has slowly formed as to trigger a gout flare.


Subject(s)
Crystallization , Gout , Uric Acid , Uric Acid/metabolism , Humans , Gout/metabolism , Gout/pathology , Biomineralization , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Arthritis, Gouty/metabolism , Arthritis, Gouty/pathology
8.
Nat Commun ; 15(1): 5705, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977710

ABSTRACT

In nature, coenzyme-independent oxidases have evolved in selective catalysis using isolated substrate-binding pockets. Single-atom nanozymes (SAzymes), an emerging type of non-protein artificial enzymes, are promising to simulate enzyme active centers, but owing to the lack of recognition sites, realizing substrate specificity is a formidable task. Here we report a metal-ligand dual-site SAzyme (Ni-DAB) that exhibited selectivity in uric acid (UA) oxidation. Ni-DAB mimics the dual-site catalytic mechanism of urate oxidase, in which the Ni metal center and the C atom in the ligand serve as the specific UA and O2 binding sites, respectively, characterized by synchrotron soft X-ray absorption spectroscopy, in situ near ambient pressure X-ray photoelectron spectroscopy, and isotope labeling. The theoretical calculations reveal the high catalytic specificity is derived from not only the delicate interaction between UA and the Ni center but also the complementary oxygen reduction at the beta C site in the ligand. As a potential application, a Ni-DAB-based biofuel cell using human urine is constructed. This work unlocks an approach of enzyme-like isolated dual sites in boosting the selectivity of non-protein artificial enzymes.


Subject(s)
Oxidation-Reduction , Urate Oxidase , Uric Acid , Substrate Specificity , Urate Oxidase/chemistry , Urate Oxidase/metabolism , Uric Acid/chemistry , Uric Acid/metabolism , Uric Acid/urine , Ligands , Humans , Nickel/chemistry , Nickel/metabolism , Binding Sites , Catalytic Domain , Catalysis , Models, Molecular , X-Ray Absorption Spectroscopy
9.
Inorg Chem ; 63(29): 13602-13612, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-38973094

ABSTRACT

Metal nanozymes have offered attractive opportunities for biocatalysis and biomedicine. However, fabricating nanozymes simultaneously possessing highly catalytic selectivity and activity remains a great challenge due to the lack of three-dimensional (3D) architecture of the catalytic pocket in natural enzymes. Here, we integrate rhodium nanocluster (RhNC), reduced graphene oxide (rGO), and protamine (PRTM, a typical arginine-rich peptide) into a composite facilely based on the single peptide. Remarkably, the PRTM-RhNC@rGO composite displays outstanding selectivity, activity, and stability for the catalytic degradation of uric acid. The reaction rate constant of the uric acid oxidation catalyzed by the PRTM-RhNC@rGO composite is about 1.88 × 10-3 s-1 (4 µg/mL), which is 37.6 times higher than that of reported RhNP (k = 5 × 10-5 s-1, 20 µg/mL). Enzyme kinetic studies reveal that the PRTM-RhNC@rGO composite exhibits a similar affinity for uric acid as natural uricase. Furthermore, the uricase-like activity of PRTM-RhNC@rGO nanozymes remains in the presence of sulfur substances and halide ions, displaying incredibly well antipoisoning abilities. The analysis of the structure-function relationship indicates the PRTM-RhNC@rGO composite features the substrate binding site near the catalytic site in a confined space contributed by 2D rGO and PRTM, resulting in the high-performance of the composite nanozyme. Based on the outstanding uricase-like activity and the interaction of PRTM and uric acid, the PRTM-RhNC@rGO composite can retard the urate crystallization significantly. The present work provides new insights into the design of metal nanozymes with suitable binding sites near catalytic sites by mimicking pocket-like structures in natural enzymes based on simple peptides, conducing to broadening the practical application of high-performance nanozymes in biomedical fields.


Subject(s)
Graphite , Rhodium , Uric Acid , Graphite/chemistry , Uric Acid/chemistry , Uric Acid/metabolism , Rhodium/chemistry , Urate Oxidase/chemistry , Urate Oxidase/metabolism , Peptides/chemistry , Peptides/pharmacology , Oxidation-Reduction , Arginine/chemistry , Metal Nanoparticles/chemistry
10.
Zhongguo Zhong Yao Za Zhi ; 49(12): 3160-3167, 2024 Jun.
Article in Chinese | MEDLINE | ID: mdl-39041076

ABSTRACT

The abnormal production and/or excretion of uric acid can lead to a disorder in uric acid metabolism, resulting in hyperuricemia, uric acid nephropathy, gouty arthritis, and other diseases related to uric acid metabolism disorder. The clinical incidence of these diseases is increasing year after year, posing a significant threat to public health. In the past, hyperuricemia and gouty arthritis were often considered different diseases, with uric acid nephropathy being a complication of hyperuricemia. However, recent research has challenged this perspective, suggesting that hyperuricemia, uric acid nephropathy, and gouty arthritis are different stages of the same disease, with urate deposition as the common pathological feature. This article offered a comprehensive overview of the current understanding of hyperuricemia, uric acid nephropathy, and gouty arthritis in both traditional Chinese medicine(TCM) and western medicine. It delved into the most up-to-date insights into the involvement of urate deposition in the pathogenesis of uric acid metabolism disorders and highlighted the dominant role of TCM in the prevention and treatment of uric acid metabolism disorders, so as to provide a reference for effective intervention strategies and drug development in uric acid metabolism disorder-related diseases.


Subject(s)
Drugs, Chinese Herbal , Hyperuricemia , Medicine, Chinese Traditional , Uric Acid , Humans , Uric Acid/metabolism , Hyperuricemia/drug therapy , Hyperuricemia/metabolism , Drugs, Chinese Herbal/therapeutic use , Arthritis, Gouty/metabolism , Arthritis, Gouty/drug therapy , Arthritis, Gouty/prevention & control , Animals
11.
Exp Parasitol ; 263-264: 108804, 2024.
Article in English | MEDLINE | ID: mdl-39019304

ABSTRACT

Research on the use of entomopathogenic nematodes (EPNs) as a potential tool for the biological control of invertebrates has been growing in recent years, including studies involving snails with One Health importance. In this study, the effect of exposure time (24 or 48 h) of Heterorhabditis bacteriophora HP88 on the activities of alanine aminotransferase (ALT) and aspartate aminotransferase (AST), as well as the concentration of total proteins, uric acid, and urea in the hemolymph of Biomphalaria glabrata, were investigated. The concentrations of these metabolic markers were measured weekly until the end of the third week after exposure. Along with a significant reduction in total protein levels, a significant increase (p < 0.01) in uric acid and urea contents in the hemolymph of B. glabrata exposed to H. bacteriophora was observed. The accumulation of urea in these mollusks could lead to deleterious effects due to its high toxicity, inducing significant cell damage. Variations in transaminase activities were also observed, with snails exposed to EPNs showing significantly higher values (p < 0.01) than individuals in the control group, both for ALT and AST. These results indicate that experimental exposure to infective juveniles of H. bacteriophora causes significant alterations in the metabolic pattern of B. glabrata, compromising the maintenance of its homeostasis. Finally, exposure for 48 h caused more damage to the planorbid in question compared to snails exposed for 24 h, suggesting that the exposure time may influence the intensity of the host's response.


Subject(s)
Alanine Transaminase , Aspartate Aminotransferases , Biomphalaria , Hemolymph , Pest Control, Biological , Rhabditoidea , Urea , Uric Acid , Animals , Biomphalaria/parasitology , Hemolymph/chemistry , Hemolymph/parasitology , Hemolymph/metabolism , Alanine Transaminase/metabolism , Aspartate Aminotransferases/metabolism , Uric Acid/metabolism , Urea/metabolism , Rhabditoidea/physiology , Proteins/metabolism , Rhabditida/physiology
12.
Psychoneuroendocrinology ; 168: 107120, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39002453

ABSTRACT

Acute physiological responses to psychosocial stressors are a potential pathway underlying racial disparities in stress-related illnesses. Uric acid (UA) is a potent antioxidant that has been linked to disparities in stress-related illnesses, and recent research has shown that UA is responsive to acute social stress. However, an examination of the relationships between the purinergic system and other commonly measured stress systems is lacking. Here, we measure and characterize associations of salivary uric acid (sUA) with markers of hypothalamic-pituitary-adrenal (HPA) axis activation, sympathetic-adreno-medullar (SAM) axis activation, and acute inflammation. A community sample of 103 African Americans (33 male, 70 female) completed the Trier Social Stress Test to induce social-evaluative threat. Passive drool collected before, during, and after the stressor task provided salivary reactivity measures of UA (sUA), cortisol, dehydroepiandrosterone sulfate (DHEAS), salivary alpha amylase (sAA - a surrogate marker of SAM activity) and C-reactive protein (sCRP). Multiple regressions revealed that total activation of cortisol, DHEAS, and sCRP were each positively associated with higher total activation of sUA. Additionally, DHEAS reactivity was positively associated with sUA reactivity. Relationships between HPA-axis markers and sUA were especially observed among younger and male participants. Overall, findings suggest potential coordination of stress systems with sUA in response to acute stress, which may further the contributions of biological stress processes to racial health disparities.


Subject(s)
Black or African American , C-Reactive Protein , Dehydroepiandrosterone Sulfate , Hydrocortisone , Hypothalamo-Hypophyseal System , Pituitary-Adrenal System , Saliva , Stress, Psychological , Urban Population , Uric Acid , Humans , Male , Female , Uric Acid/metabolism , Uric Acid/analysis , Saliva/chemistry , Saliva/metabolism , Adult , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Hydrocortisone/metabolism , Hydrocortisone/analysis , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Middle Aged , Dehydroepiandrosterone Sulfate/metabolism , Dehydroepiandrosterone Sulfate/analysis , C-Reactive Protein/metabolism , C-Reactive Protein/analysis , Salivary alpha-Amylases/metabolism , Salivary alpha-Amylases/analysis , Biomarkers/metabolism , Young Adult
13.
Nat Commun ; 15(1): 6463, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39085241

ABSTRACT

Hyperuricemia is associated with an increased risk of gout, hypertension, diabetes, and cardiovascular diseases. Most mammals maintain normal serum uric acid (SUA) via urate oxidase (Uox), an enzyme that metabolizes poorly-soluble UA to highly-soluble allantoin. In contrast, Uox became a pseudogene in humans and apes over the long course of evolution. Here we demonstrate an atavistic strategy for treating hyperuricemia based on endogenous expression of Uox in hepatocytes mediated by mRNA (mUox) loaded with an ionizable lipid nanoparticle termed iLAND. mUox@iLAND allows effective transfection and protein expression in vitro. A single dose of mUox@iLAND lowers SUA levels for several weeks in two female murine models, including a novel long-lasting model, which is also confirmed by metabolomics analysis. Together with the excellent safety profiles observed in vivo, the proposed mRNA agent demonstrates substantial potential for hyperuricemia therapy and the prevention of associated conditions.


Subject(s)
Hyperuricemia , Liposomes , RNA, Messenger , Urate Oxidase , Uric Acid , Hyperuricemia/drug therapy , Hyperuricemia/genetics , Hyperuricemia/metabolism , Animals , RNA, Messenger/metabolism , RNA, Messenger/genetics , Urate Oxidase/metabolism , Urate Oxidase/genetics , Female , Mice , Humans , Uric Acid/metabolism , Uric Acid/blood , Liposomes/chemistry , Nanoparticles/chemistry , Hepatocytes/metabolism , Disease Models, Animal , Mice, Inbred C57BL
14.
Front Biosci (Landmark Ed) ; 29(6): 222, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38940057

ABSTRACT

BACKGROUND: Persistent hyperuricemia can lead to the generation and deposition of monosodium urate (MSU) crystals. This can trigger gouty arthritis (GA), which in turn induces inflammation. Activation of the Nod-like receptor pyrin domain containing 3 (NLRP3) inflammasome plays a critical role in the onset and progression of GA. Autophagy may have a dual effect on GA with regard to the NLRP3 inflammasome. Therefore, the present study aimed to gain a deeper comprehension of the interaction between autophagy and NLRP3 inflammasome activation is imperative for developing more efficacious treatments for GA. METHODS: Peripheral blood monocytes (PBMCs) were first isolated from GA patients and healthy controls and underwent bulk RNA sequencing analysis. Overexpression and knockdown of dual specificity phosphatase 1 (DUSP1) was performed in THP-1 monocytes to investigate its role in the immune response and mitochondrial damage. The luciferase assay and Western blot analysis were used to study the interaction between autophagy and NLRP3 inflammasome activation. RESULTS: Bulk RNA sequencing analysis showed significant upregulation of DUSP1 expression in PBMCs from GA patients compared to healthy controls. This result was subsequently verified by reverse transcription quantitative polymerase chain reaction (RT-qPCR). DUSP1 expression in human THP-1 monocytes was also shown to increase after MSU treatment. Downregulation of DUSP1 expression increased the secretion of inflammatory cytokines after MSU treatment, whereas the overexpression of DUSP1 decreased the secretion levels. Lipopolysaccharides (LPS) combined with adenosine-triphosphate (ATP) led to mitochondrial damage, which was rescued by overexpressing DUSP1. DUSP1 overexpression further increased the level of autophagy following MSU treatment, whereas downregulation of DUSP1 decreased autophagy. Treatment with the autophagy inhibitor 3-Methyladenine (3-MA) restored inflammatory cytokine secretion levels in the DUSP1 overexpression group. MSU caused pronounced pathological ankle swelling in vivo. However, DUSP1 overexpression significantly mitigated this phenotype, accompanied by significant downregulation of inflammatory cytokine secretion levels in the joint tissues. CONCLUSIONS: This study revealed a novel function and mechanism for DUSP1 in promoting autophagy to mitigate the MSU-induced immune response in GA. This finding suggests potential diagnostic biomarkers and anti-inflammatory targets for more effective GA therapy.


Subject(s)
Arthritis, Gouty , Autophagy , Dual Specificity Phosphatase 1 , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Uric Acid , Humans , Autophagy/drug effects , Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 1/metabolism , Arthritis, Gouty/genetics , Arthritis, Gouty/metabolism , Arthritis, Gouty/immunology , Arthritis, Gouty/chemically induced , Uric Acid/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Inflammasomes/metabolism , Inflammasomes/immunology , THP-1 Cells , Male , Monocytes/metabolism , Monocytes/immunology , Monocytes/drug effects , Case-Control Studies , Female , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Middle Aged
15.
Arch Biochem Biophys ; 758: 110078, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38944139

ABSTRACT

About 140 million people worldwide live at an altitude above 2500 m. Studies have showed an increase of the incidence of hyperuricemia among plateau populations, but little is known about the possible mechanisms. This study aims to assess the effects of high altitude on hyperuricemia and explore the corresponding mechanisms at the histological, inflammatory and molecular levels. This study finds that intermittent hypobaric hypoxia (IHH) exposure results in an increase of serum uric acid level and a decrease of uric acid clearance rate. Compared with the control group, the IHH group shows significant increases in hemoglobin concentration (HGB) and red blood cell counts (RBC), indicating that high altitude hyperuricemia is associated with polycythemia. This study also shows that IHH exposure induces oxidative stress, which causes the injury of liver and renal structures and functions. Additionally, altered expressions of organic anion transporter 1 (OAT1) and organic cation transporter 1 (OCT1) of kidney have been detected in the IHH exposed rats. The adenosine deaminase (ADA) expression levels and the xanthione oxidase (XOD) and ADA activity of liver of the IHH exposure group have significantly increased compared with those of the control group. Furthermore, the spleen coefficients, IL-2, IL-1ß and IL-8, have seen significant increases among the IHH exposure group. TLR/MyD88/NF-κB pathway is activated in the process of IHH induced inflammatory response in joints. Importantly, these results jointly show that IHH exposure causes hyperuricemia. IHH induced oxidative stress along with liver and kidney injury, unusual expression of the uric acid synthesis/excretion regulator and inflammatory response, thus suggesting a potential mechanism underlying IHH-induced hyperuricemia.


Subject(s)
Hyperuricemia , Hypoxia , Kidney , Liver , Oxidative Stress , Hyperuricemia/metabolism , Animals , Male , Rats , Liver/metabolism , Liver/pathology , Hypoxia/metabolism , Hypoxia/complications , Kidney/metabolism , Kidney/pathology , Altitude , Uric Acid/blood , Uric Acid/metabolism , Rats, Sprague-Dawley , Xanthine Oxidase/metabolism , Altitude Sickness/metabolism , Altitude Sickness/complications , Altitude Sickness/physiopathology
16.
Theranostics ; 14(8): 3082-3103, 2024.
Article in English | MEDLINE | ID: mdl-38855180

ABSTRACT

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


Subject(s)
Alginates , Arthritis, Gouty , Disease Models, Animal , Inflammation , Oligosaccharides , Animals , Arthritis, Gouty/drug therapy , Arthritis, Gouty/metabolism , Mice , Oligosaccharides/pharmacology , Alginates/pharmacology , Inflammation/drug therapy , Inflammation/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Male , Arthralgia/drug therapy , Arthralgia/metabolism , Uric Acid/metabolism , Mice, Inbred C57BL , Anti-Inflammatory Agents/pharmacology , Inflammasomes/metabolism , Inflammasomes/drug effects , Ankle Joint/pathology , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects
17.
Yakugaku Zasshi ; 144(6): 659-674, 2024.
Article in Japanese | MEDLINE | ID: mdl-38825475

ABSTRACT

Serum urate levels are determined by the balance between uric acid production and uric acid excretion capacity from the kidneys and intestinal tract. Dysuricemia, including hyperuricemia and hypouricemia, develops when the balance shifts towards an increase or a decrease in the uric acid pool. Hyperuricemia is mostly a multifactorial genetic disorder involving several disease susceptibility genes and environmental factors. Hypouricemia, on the other hand, is caused by genetic abnormalities. The main genes involved in dysuricemia are xanthine oxidoreductase, an enzyme that produces uric acid, and the urate transporters urate transporter 1/solute carrier family 22 member 12 (URAT1/SLC22A12), glucose transporter 9/solute carrier family 2 member 9 (GLUT9/SLC2A9) and ATP binding cassette subfamily G member 2 (ABCG2). Deficiency of xanthine oxidoreductase results in xanthinuria, a rare disease with marked hypouricemia. Xanthinuria can be due to a single deficiency of xanthine oxidoreductase or in combination with aldehyde oxidase deficiency as well. The latter is caused by a deficiency in molybdenum cofactor sulfurase, which is responsible for adding sulphur atoms to the molybdenum cofactor required for xanthine oxidoreductase and aldehyde oxidase to exert their action. URAT1/SLC22A12 and GLUT9/SLC2A9 are involved in urate reabsorption and their deficiency leads to renal hypouricemia, a condition that is common in Japanese due to URAT1/SLC22A12 deficiency. On the other hand, ABCG2 is involved in the secretion of urate, and many Japanese have single nucleotide polymorphisms that result in its reduced function, leading to hyperuricemia. In particular, severe dysfunction of ABCG2 leads to hyperuricemia with reduced extrarenal excretion.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2 , Glucose Transport Proteins, Facilitative , Hyperuricemia , Neoplasm Proteins , Organic Anion Transporters , Uric Acid , Xanthine Dehydrogenase , Humans , Hyperuricemia/etiology , Hyperuricemia/metabolism , Hyperuricemia/genetics , Uric Acid/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Organic Anion Transporters/metabolism , Organic Anion Transporters/genetics , Glucose Transport Proteins, Facilitative/metabolism , Glucose Transport Proteins, Facilitative/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Xanthine Dehydrogenase/metabolism , Xanthine Dehydrogenase/genetics , Xanthine Dehydrogenase/deficiency , Animals , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Renal Tubular Transport, Inborn Errors/genetics , Renal Tubular Transport, Inborn Errors/etiology , Renal Tubular Transport, Inborn Errors/metabolism , Urinary Calculi/etiology , Urinary Calculi/metabolism , Urinary Calculi/genetics , Metabolism, Inborn Errors
18.
Ter Arkh ; 96(5): 447-452, 2024 Jun 03.
Article in Russian | MEDLINE | ID: mdl-38829804

ABSTRACT

This article examines the role of uric acid (UA) in cognitive changes and neurodegeneration, focusing on its functions as an antioxidant and prooxidant. Research suggests that changes in serum UA levels may be associated with the development or delay of cognitive impairment, especially in the context of neurodegenerative diseases such as Alzheimer's disease. It was revealed that there is a relationship between the level of UA and the dynamics of cognitive functions, indicating the potential neuroprotective properties of UA. Particular attention is paid to the balance between the antioxidant and prooxidant properties of UA, which may play a key role in protecting neurons from damage. However, research results are not clear-cut, highlighting the need for further research to more fully understand the role of UA in cognitive processes. Determining the optimal serum UA level may be an important step in developing strategies for the prevention and treatment of cognitive impairment associated with neurodegeneration. Overall, these studies advance the understanding of the mechanisms underlying the interaction between uric acid metabolism and brain health.


Subject(s)
Neurodegenerative Diseases , Uric Acid , Humans , Uric Acid/blood , Uric Acid/metabolism , Neurodegenerative Diseases/physiopathology , Neurodegenerative Diseases/metabolism , Cognition Disorders/etiology , Cognition Disorders/prevention & control , Cognition Disorders/physiopathology , Antioxidants , Alzheimer Disease/physiopathology , Alzheimer Disease/metabolism , Brain/metabolism , Brain/physiopathology , Oxidative Stress/physiology
19.
Nat Commun ; 15(1): 5039, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38866775

ABSTRACT

Urate, the physiological form of uric acid and a potent antioxidant in serum, plays a pivotal role in scavenging reactive oxygen species. Yet excessive accumulation of urate, known as hyperuricemia, is the primary risk factor for the development of gout. The high-capacity urate transporter GLUT9 represents a promising target for gout treatment. Here, we present cryo-electron microscopy structures of human GLUT9 in complex with urate or its inhibitor apigenin at overall resolutions of 3.5 Å and 3.3 Å, respectively. In both structures, GLUT9 exhibits an inward open conformation, wherein the substrate binding pocket faces the intracellular side. These structures unveil the molecular basis for GLUT9's substrate preference of urate over glucose, and show that apigenin acts as a competitive inhibitor by occupying the substrate binding site. Our findings provide critical information for the development of specific inhibitors targeting GLUT9 as potential therapeutics for gout and hyperuricemia.


Subject(s)
Apigenin , Cryoelectron Microscopy , Glucose Transport Proteins, Facilitative , Uric Acid , Humans , Glucose Transport Proteins, Facilitative/metabolism , Glucose Transport Proteins, Facilitative/antagonists & inhibitors , Glucose Transport Proteins, Facilitative/chemistry , Uric Acid/metabolism , Uric Acid/chemistry , Apigenin/pharmacology , Apigenin/chemistry , Binding Sites , Protein Binding , Hyperuricemia/drug therapy , Hyperuricemia/metabolism , Models, Molecular , Gout/drug therapy , Gout/metabolism , HEK293 Cells
SELECTION OF CITATIONS
SEARCH DETAIL