Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.842
Filter
1.
Med J Malaysia ; 79(4): 457-463, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39086344

ABSTRACT

INTRODUCTION: Hyperuricaemia is common in essential hypertension with varying results in different populations. This study sought to ascertain the association between serum uric acid levels and essential hypertension in Hospital Universiti Sains Malaysia (HUSM). MATERIALS AND METHODS: A case-control study design involving 132 subjects (88 subjects of hypertension patients for case group and 44 subjects for control group) aged 18 to 40 years old of both genders was conducted at HUSM primary care clinic and physician clinic from May 2020 to May 2021. Blood samples were collected from each of the case and control subjects and analysed for serum uric acid, urea, creatinine, total cholesterol, triglycerides, LDL and HDL on chemical analyser Architect c8000. The data were analysed by using SPSS Statistics 26.0 version. RESULTS: The proportion of subjects with hyperuricaemia in the case group was 48.9%. A significant difference in the uric acid levels between the case group (390.64±92.65µmol/L) and control group (352.09±86.07µmol/L), (p<0.05) was observed. There was no significant difference in the serum uric acid mean ± SD based on the duration of hypertension (<5 years and ≥5 years), (p=0.331) and stages of hypertension (p>0.05). In case group, significant correlations were established between uric acid and triglycerides (r=0.255, p<0.05), uric acid and HDL (r= -0.223, p<0.05), uric acid and urea (r=0.299, p<0.05), uric acid and creatinine (r=0.486, p<0.01). No correlation among uric acid and total cholesterol levels (p>0.05), uric acid and LDL (p>0.05). Serum uric acid was a vital variable in developing hypertension (p<0.05) but not when adapted for age and body mass index (BMI) (p>0.05). CONCLUSION: Serum uric acid was significantly elevated in essential hypertension. The significant associations were established between uric acid and triglycerides, HDL, urea and creatinine in essential hypertension. Serum uric acid was a vital variable to develop hypertension, but the association was weakened by other co-founders as age and BMI. A large-scale population-based study is required to truly conclude the association between serum uric acid levels and essential hypertension in our population.


Subject(s)
Essential Hypertension , Hyperuricemia , Uric Acid , Humans , Uric Acid/blood , Male , Malaysia , Female , Adult , Case-Control Studies , Essential Hypertension/blood , Young Adult , Hyperuricemia/blood , Adolescent , Creatinine/blood , Urea/blood
2.
Front Immunol ; 15: 1386939, 2024.
Article in English | MEDLINE | ID: mdl-39100670

ABSTRACT

Objective: This study aimed to evaluate the role of absent in melanoma 2 (AIM2) inflammasome-mediated pyroptosis in the pathogenesis of acute gouty arthritis (AGA) and asymptomatic hyperuricemia(AHU). Methods: A cohort of 30 AGA patients, 30 AHU individuals, and 30 healthy controls (HC) was assembled. Demographic and biochemical data, along with blood samples, were collected. Serum double-stranded DNA (dsDNA) levels were quantified using a fluorescent assay. Transcriptomic and proteomic analysis of AIM2, Caspase-1, GSDMD, IL-1ß, and IL-18 in peripheral blood mononuclear cells was performed using qRT-PCR and Western blot. Enzyme-linked immunosorbent assay (ELISA) was employed to measure serum IL-1ß and IL-18. Spearman correlation analysis was utilized to assess relationships between variables. Results: Both AGA and AHU groups demonstrated elevated metabolic indicators and serum levels of dsDNA, IL-1ß, and IL-18 compared to the HC group. AGA patients exhibited higher inflammatory markers than the AHU group. In the AGA group, there was a significant increase in the mRNA and protein levels of AIM2, Caspase-1, GSDMD, IL-1ß, and IL-18 (P<0.05 to P<0.001). The AHU group showed higher AIM2, Caspase-1, GSDMD, and IL-18 mRNA levels than the HC group (P<0.001 to P<0.01), with a non-significant increase in AIM2, GSDMD, and IL-1ß proteins (P>0.05). In contrast, Caspase-1 and IL-18 proteins were significantly higher in the AHU group (P<0.05). Notable correlations were observed between AIM2 protein expression and levels of Caspase-1 and GSDMD in both AGA and AHU groups. In the AGA group, AIM2 protein correlated with IL-1ß, but not in the AHU group. The AIM2 protein in the AHU group was positively associated with IL-18, with no such correlation in the AGA group. Conclusion: AIM2 inflammasome may play a role in the inflammatory processes of AGA and AHU and that its activation may be related to the pyroptosis pathway.


Subject(s)
Arthritis, Gouty , DNA-Binding Proteins , Hyperuricemia , Inflammasomes , Pyroptosis , Humans , Male , Inflammasomes/metabolism , Arthritis, Gouty/immunology , Arthritis, Gouty/blood , Arthritis, Gouty/metabolism , Middle Aged , Hyperuricemia/blood , Hyperuricemia/immunology , Female , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Adult , Interleukin-18/blood , Aged , Case-Control Studies , Biomarkers/blood , Caspase 1/metabolism
3.
PLoS One ; 19(8): e0306025, 2024.
Article in English | MEDLINE | ID: mdl-39088565

ABSTRACT

The relationship between molybdenum and kidney-related disease outcomes, including hyperuricemia, is not well investigated. This study aims to determine whether molybdenum and its antioxidative property are associated with systemic inflammation and kidney-related disease parameters including hyperuricemia. Urinary molybdenum's epidemiological relationship to hyperuricemia and kidney-disease related outcomes was evaluated in 15,370 adult participants in the National Health and Nutrition Examination Survey (NHANES) collected between 1999 and 2016. Individuals' urinary molybdenum levels were corrected to their urinary creatinine concentrations. The association between urinary molybdenum-to-creatinine ratio and kidney-disease related outcomes were assessed by multivariable linear and logistic regression analyses, adjusting for covariates including age, sex, ethnicity, diabetes mellitus, hypertension, body mass index, and estimated glomerular filtration rate. Antimony and tungsten were used as control trace metals. Experimentally, HK-2 cell was used to assess molybdenum's antioxidative properties. HK-2 cells were challenged with H2O2-induced oxidative stress. Oxidative stress was measured using a fluorescent microplate assay for reactive oxygen species (ROS) and antioxidation levels were assessed by measuring the expression of manganese superoxide dismutase. In the adult NHANES population, urinary molybdenum-to-creatinine ratio was significantly associated with decreased serum uric acid (ß, -0.119; 95% CI, -0.148 to -0.090) concentrations, and decreased prevalence of hyperuricemia (OR, 0.73; 95% CI, 0.64-0.83) and gout (OR, 0.71; 95% CI, 0.52-0.94). Higher urinary molybdenum levels were associated with lower levels of systemic oxidative stress (gamma-glutamyltransferase levels; ß, -0.052; 95% CI, -0.067 to -0.037) and inflammation (C-reactive protein levels; ß, -0.184; 95% CI, -0.220 to -0.148). In HK-2 cells under H2O2-induced oxidative stress, molybdenum upregulated manganese superoxide dismutase expression and decreased oxidative stress. Urinary molybdenum levels are associated with decreased prevalence of hyperuricemia and gout in adult population. Molybdenum's antioxidative properties might have acted as an important mechanism for the reduction of systemic inflammation, ROS, and uric acid levels.


Subject(s)
Antioxidants , Hyperuricemia , Molybdenum , Oxidative Stress , Humans , Hyperuricemia/epidemiology , Molybdenum/urine , Adult , Female , Antioxidants/metabolism , Male , Middle Aged , Prevalence , Oxidative Stress/drug effects , Creatinine/urine , Creatinine/blood , Reactive Oxygen Species/metabolism , Nutrition Surveys , Cell Line , Uric Acid/blood , Uric Acid/urine
4.
Sci Rep ; 14(1): 18466, 2024 08 09.
Article in English | MEDLINE | ID: mdl-39122851

ABSTRACT

The bidirectional effect of hyperuricemia on chronic kidney disease (CKD) underscores the importance of hyperuricemia as a risk factor for CKD. We evaluated the effect of hyperuricemia on the presence and development of CKD after considering genetic background by calculating polygenic risk scores (PRSs). We employed genome-wide association study summary statistics-excluding the United Kingdom Biobank (UKB) datasets among published CKD Gen Consortium papers-to calculate the PRSs for CKD in white background subjects. To validate PRS performance, we divided the UKB into two datasets to validate and test the data. We used logistic regression analysis to evaluate the association between hyperuricemia and CKD, and performed Kaplan-Meier survival analysis exclusively for subjects with available follow-up data. In total, 438,253 clinical data and 4,307,940 single nucleotide polymorphisms from 459,155 samples were included. We observed a significant positive association between PRS and CKD and the presence and development of CKD. Hyperuricemia significantly increased CKD risk (adjusted odds ratio 1.55, 95% confidence interval 1.48-1.61). The impact of hyperuricemia on CKD was maintained irrespective of PRS range. In addition, negative interaction between hyperuricemia and PRS for CKD was found. Survival analysis indicates that the presence of hyperuricemia significantly increased the risk of CKD development. The PRS for CKD thoroughly reflects the risk of CKD development. Hyperuricemia is a significant indicator of CKD risk, even after incorporating the genetic risk score for CKD. Irrespective of genetic risk, patients with a prospective risk of developing CKD require uric acid monitoring and management.


Subject(s)
Genetic Predisposition to Disease , Genome-Wide Association Study , Hyperuricemia , Polymorphism, Single Nucleotide , Renal Insufficiency, Chronic , Humans , Hyperuricemia/genetics , Hyperuricemia/complications , Renal Insufficiency, Chronic/genetics , Male , Female , Middle Aged , Risk Factors , Cohort Studies , United Kingdom/epidemiology , Aged , Adult , Multifactorial Inheritance
5.
World J Gastroenterol ; 30(29): 3488-3510, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39156502

ABSTRACT

BACKGROUND: Hyperuricemia (HUA) is a public health concern that needs to be solved urgently. The lyophilized powder of Poecilobdella manillensis has been shown to significantly alleviate HUA; however, its underlying metabolic regulation remains unclear. AIM: To explore the underlying mechanisms of Poecilobdella manillensis in HUA based on modulation of the gut microbiota and host metabolism. METHODS: A mouse model of rapid HUA was established using a high-purine diet and potassium oxonate injections. The mice received oral drugs or saline. Additionally, 16S rRNA sequencing and ultra-high performance liquid chromatography with quadrupole time-of-flight mass spectrometry-based untargeted metabolomics were performed to identify changes in the microbiome and host metabolome, respectively. The levels of uric acid transporters and epithelial tight junction proteins in the renal and intestinal tissues were analyzed using an enzyme-linked immunosorbent assay. RESULTS: The protein extract of Poecilobdella manillensis lyophilized powder (49 mg/kg) showed an enhanced anti-trioxypurine ability than that of allopurinol (5 mg/kg) (P < 0.05). A total of nine bacterial genera were identified to be closely related to the anti-trioxypurine activity of Poecilobdella manillensis powder, which included the genera of Prevotella, Delftia, Dialister, Akkermansia, Lactococcus, Escherichia_Shigella, Enterococcus, and Bacteroides. Furthermore, 22 metabolites in the serum were found to be closely related to the anti-trioxypurine activity of Poecilobdella manillensis powder, which correlated to the Kyoto Encyclopedia of Genes and Genomes pathways of cysteine and methionine metabolism, sphingolipid metabolism, galactose metabolism, and phenylalanine, tyrosine, and tryptophan biosynthesis. Correlation analysis found that changes in the gut microbiota were significantly related to these metabolites. CONCLUSION: The proteins in Poecilobdella manillensis powder were effective for HUA. Mechanistically, they are associated with improvements in gut microbiota dysbiosis and the regulation of sphingolipid and galactose metabolism.


Subject(s)
Disease Models, Animal , Gastrointestinal Microbiome , Hyperuricemia , Leeches , Animals , Hyperuricemia/drug therapy , Hyperuricemia/blood , Hyperuricemia/microbiology , Gastrointestinal Microbiome/drug effects , Mice , Male , Leeches/microbiology , Uric Acid/blood , Kidney/drug effects , Kidney/metabolism , Kidney/microbiology , Metabolomics/methods , RNA, Ribosomal, 16S/genetics , Humans , Dysbiosis , Metabolome/drug effects
6.
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
7.
Nutr J ; 23(1): 89, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39123196

ABSTRACT

BACKGROUND: We aimed to probe the association of serum 25-hydroxyvitamin D [25(OH)D] concentrations with all-cause and cause-specific mortality among patients with gout and hyperuricemia (HUA). METHODS: The study included 1169 gout patients and 7029 HUA patients from the National Health and Nutrition Examination Survey (NHANES) 2007-2018 and 2001-2018, respectively. The association between serum 25(OH)D and mortality was evaluated by Cox proportional hazard and restricted cubic spline models. RESULTS: Among participants with gout and HUA, the weighted mean concentrations of serum 25(OH)D were 71.49 ± 30.09 nmol/L and 64.81 ± 26.92 nmol/L, respectively. Vitamin D deficiency occurred in 29.68% of gout patients and 37.83% of HUA patients. During 6783 person-years of follow-up among gout patients, 248 all-cause deaths occurred, among which 76 died from cardiovascular disease (CVD) and 49 died from cancer. 1375 HUA patients were recorded for all-cause mortality during 59,859 person-years of follow-up, including 427 CVD deaths and 232 cancer deaths. After multifactorial adjustment, per one-unit increment in natural log-transformed 25(OH)D was associated with lower risk of 55% all-cause mortality and 61% CVD mortality among gout patients, and a 45% reduced risk of cancer mortality among HUA patients. Restricted cubic splines showed a U-shaped relationship with all-cause and CVD mortality among HUA patients, with inflection points of 72.7 nmol/L and 38.0 nmol/L, respectively. The results were robust in subgroup and sensitivity analyses. CONCLUSIONS: Serum 25(OH)D was negatively linearly correlated with mortality among gout patients, whereas U-shaped correlated with mortality in HUA patients. These results indicate that adequate vitamin D status could prevent premature death.


Subject(s)
Cause of Death , Gout , Hyperuricemia , Nutrition Surveys , Vitamin D , Humans , Gout/blood , Gout/mortality , Gout/complications , Hyperuricemia/blood , Hyperuricemia/mortality , Hyperuricemia/complications , Vitamin D/analogs & derivatives , Vitamin D/blood , Male , Female , Middle Aged , Nutrition Surveys/statistics & numerical data , Adult , Aged , Cardiovascular Diseases/mortality , Cardiovascular Diseases/blood , Neoplasms/mortality , Neoplasms/blood , Neoplasms/complications , Vitamin D Deficiency/blood , Vitamin D Deficiency/complications , Vitamin D Deficiency/mortality , Proportional Hazards Models
8.
Front Endocrinol (Lausanne) ; 15: 1415459, 2024.
Article in English | MEDLINE | ID: mdl-39135624

ABSTRACT

Objectives: This study aimed to explore the synergistic interaction effect between hyperuricemia and hypertension towards chronic kidney disease in patients with type 2 diabetes. Methods: This research originates from a cross-sectional study performed in Zhejiang Province, Eastern China, between March and November 2018. The correlation between serum uric acid levels and the risk of chronic kidney disease was assessed using a restricted cubic spline model. An unconditional multivariable logistic regression model, along with an interaction table, was utilized to explore the potential interaction effect of hyperuricemia and hypertension towards chronic kidney disease. Results: 1,756 patients with type 2 diabetes were included in this study, the prevalence of chronic kidney disease (CKD) was 27.62% in this population. A U-shaped non-linear pattern emerged correlating serum uric acid (SUA) levels and CKD risk, indicating that both low and high SUA levels were linked to an increased CKD risk. This risk achieved its lowest point (nadir) at SUA approximately equals to 285µmol/L (p for trend <0.05). Once adjustments for age, gender, education level, abnormal fasting plasma glucose (FPG), abnormal hemoglobin A1c (HbA1c), abnormal total cholesterol (TC), abnormal high-density lipoprotein cholesterol (HDL-C), alcohol consumption and duration of diabetes were factored in, it was found that patients with both hyperuricemia and hypertension demonstrated a 5.42-fold (95% CI: 3.72-7.90) increased CKD risk compared to the reference group. The additive interaction between hyperuricemia and hypertension was statistically significant, as manifested by the following values: a relative excess risk due to interaction (RERI) of 2.57 (95% CI: 0.71-4.71), an attributable proportion due to interaction (AP) of 0.47 (95% CI: 0.14-0.64), and a synergy index (SI) of 2.39 (95% CI: 1.24-4.58). In contrast, there was no significant interaction effect in multiplicative scale. Conclusion: Hyperuricemia and hypertension may contribute additively to CKD, beyond their isolated impacts. Evaluating the risk of CKD in type 2 diabetes patients necessitates considering this potential interaction.


Subject(s)
Diabetes Mellitus, Type 2 , Hypertension , Hyperuricemia , Renal Insufficiency, Chronic , Uric Acid , Humans , Hyperuricemia/epidemiology , Hyperuricemia/blood , Hyperuricemia/complications , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/blood , Cross-Sectional Studies , Male , Female , Renal Insufficiency, Chronic/epidemiology , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/complications , Middle Aged , Hypertension/epidemiology , Hypertension/blood , Hypertension/complications , China/epidemiology , Aged , Uric Acid/blood , Risk Factors , Adult , Prevalence
9.
Int J Mol Sci ; 25(15)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39126116

ABSTRACT

Plantaginis semen is the dried ripe seed of Plantago asiatica L. or Plantago depressa Willd., which has a long history in alleviating hyperuricemia (HUA) and chronic kidney diseases. While the major chemical ingredients and mechanism remained to be illustrated. Therefore, this work aimed to elucidate the chemicals and working mechanisms of PS for HUA. UPLC-QE-Orbitrap-MS was applied to identify the main components of PS in vitro and in vivo. RNA sequencing (RNA-seq) was conducted to explore the gene expression profile, and the genes involved were further confirmed by real-time quantitative PCR (RT-qPCR). A total of 39 components were identified from PS, and 13 of them were detected in the rat serum after treating the rat with PS. The kidney tissue injury and serum uric acid (UA), xanthine oxidase (XOD), and cytokine levels were reversed by PS. Meanwhile, renal urate anion transporter 1 (Urat1) and glucose transporter 9 (Glut9) levels were reversed with PS treatment. RNA-seq analysis showed that the PPAR signaling pathway; glycine, serine, and threonine metabolism signaling pathway; and fatty acid metabolism signaling pathway were significantly modified by PS treatment. Further, the gene expression of Slc7a8, Pck1, Mgll, and Bhmt were significantly elevated, and Fkbp5 was downregulated, consistent with RNA-seq results. The PPAR signaling pathway involved Pparα, Pparγ, Lpl, Plin5, Atgl, and Hsl were elevated by PS treatment. URAT1 and PPARα proteins levels were confirmed by Western blotting. In conclusion, this study elucidates the chemical profile and working mechanisms of PS for prevention and therapy of HUA and provides a promising traditional Chinese medicine agency for HUA prophylaxis.


Subject(s)
Hyperuricemia , Oxonic Acid , Plantago , Hyperuricemia/drug therapy , Hyperuricemia/metabolism , Animals , Rats , Oxonic Acid/adverse effects , Male , Plantago/chemistry , Uric Acid/blood , Plant Extracts/pharmacology , Kidney/metabolism , Kidney/drug effects , Rats, Sprague-Dawley , Signal Transduction/drug effects , Organic Cation Transport Proteins/metabolism , Organic Cation Transport Proteins/genetics , Organic Anion Transporters/metabolism , Organic Anion Transporters/genetics , Xanthine Oxidase/metabolism
10.
Front Endocrinol (Lausanne) ; 15: 1424207, 2024.
Article in English | MEDLINE | ID: mdl-39140032

ABSTRACT

Background: Prior research has highlighted the association between uric acid (UA) and the activation of the renin-angiotensin-aldosterone system (RAAS). However, the specific relationship between aldosterone, the RAAS's end product, and UA-related diseases remains poorly understood. This study aims to clarify the impact of aldosterone on the development and progression of hyperuricemia and gout in hypertensive patients. Methods: Our study involved 34534 hypertensive participants, assessing plasma aldosterone concentration (PAC)'s role in UA-related diseases, mainly hyperuricemia and gout. We applied multiple logistic regression to investigate the impact of PAC and used restricted cubic splines (RCS) for examining the dose-response relationship between PAC and these diseases. To gain deeper insights, we conducted threshold analyses, further clarifying the nature of this relationship. Finally, we undertook subgroup analyses to evaluate PAC's effects across diverse conditions and among different subgroups. Results: Multivariate logistic regression analysis revealed a significant correlation between the occurrence of hyperuricemia and gout and the elevation of PAC levels. Compared to the first quartile (Q1) group, groups Q2, Q3, and Q4 all exhibited a significantly increased risk of occurrence. Moreover, the conducted RCS analysis demonstrated a significant nonlinear dose-response relationship, especially when PAC was greater than 14 ng/dL, with a further increased risk of hyperuricemia and gout. Finally, comprehensive subgroup analyses consistently reinforced these findings. Conclusion: This study demonstrates a close association between elevated PAC levels and the development of UA-related diseases, namely hyperuricemia and gout, in hypertensive patients. Further prospective studies are warranted to confirm and validate this relationship.


Subject(s)
Aldosterone , Gout , Hypertension , Hyperuricemia , Humans , Hyperuricemia/blood , Hyperuricemia/complications , Gout/blood , Gout/epidemiology , Gout/complications , Male , Aldosterone/blood , Hypertension/blood , Hypertension/complications , Middle Aged , Female , Aged , Uric Acid/blood , Renin-Angiotensin System/physiology , Adult
11.
Ren Fail ; 46(2): 2387429, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39132829

ABSTRACT

Objectives: To investigate the role of the intestinal flora and metabolites in the development of hyperuricemic renal injury in chronic kidney disease (CKD).Methods: Unilaterally nephrectomized mice were fed with adenine and potassium oxonate for 9 weeks. HE staining combined with plasma biochemical indicators was used to evaluate renal pathological and functional changes. We conducted 16S rRNA sequencing and untargeted metabolomics on feces and plasma samples to reveale changes in intestinal microbiota and metabolites.Result: Our analysis revealed significant differences in 15 bacterial genera, with 7 being upregulated and 8 being downregulated. Furthermore, metabolomic analysis revealed changes in the distribution of amino acid and biotin metabolites in basic metabolic pathways in both feces and serum. Specifically, differentially abundant metabolites in feces were associated primarily with histidine metabolism; the biosynthesis of phenylalanine, tyrosine, and tryptophan; and tyrosine metabolism. In plasma, the differentially abundant metabolites were involved in multiple metabolic pathways, including aminoacyl-tRNA biosynthesis; glycine, serine, and threonine amino acid metabolism; valine, leucine, and isoleucine biosynthesis; tyrosine biosynthesis and metabolism; biotin metabolism; and taurine and hypotaurine metabolism. Furthermore, correlation analysis revealed that Akkermansia, UCG-005, Lachnospiraceae_NK4A136_group, Lactococcus, and Butymonas were associated with various differentially abundant metabolites as well as renal function, oxidative stress, and mitophagy. The changes in the intestinal flora observed in hyperuricemia may lead to imbalances in amino acid and biotin metabolism in both the intestine and host, ultimately affecting oxidative stress and mitophagy in mice and accelerating the progression of CKD.Conclusion: Our findings provide insights into a potential pathogenic mechanism by which hyperuricemia exacerbates renal injury in mice with renal insufficiency. Understanding these pathways may offer new therapeutic strategies for managing hyperuricemic renal injury in CKD patients.


Subject(s)
Disease Models, Animal , Gastrointestinal Microbiome , Hyperuricemia , Animals , Hyperuricemia/metabolism , Mice , Male , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/microbiology , Metabolomics/methods , Feces/microbiology , RNA, Ribosomal, 16S , Mice, Inbred C57BL , Kidney/metabolism , Kidney/pathology
13.
Sci Rep ; 14(1): 16428, 2024 07 16.
Article in English | MEDLINE | ID: mdl-39013961

ABSTRACT

Studies investigating the relationship between dietary vitamin B1 intake and risk of Hyperuricemia (HU) are scarce, the present study aimed to examine the association of dietary vitamin B1 intake and HU among adults. This cross-sectional study included 5750 adults whose data derived from National Health and Nutrition Examination Survey (NHANES) from March 2017 to March 2020. The dietary intake of vitamin B1 was assessed using 24-h dietary recall interviews. The characteristics of study participants were grouped into five levels according to the levels of vitamin B1 quintile. Multivariate logistic regression analysis was used to estimate the odds ratio (OR) and 95% confidence interval (CI) of HU, according to the vitamin B1 intake quintile for male and female separately. The dose-response relationship was determined by the restricted cubic spline (RCS). Smoothed curve fitting was used to assess serum uric acid concentration versus dietary vitamin B1 intake in the study population. The prevalence of hyperuricemia was 18.90% (20.15% and 17.79% for males and females, respectively) in the United States from March 2017 to March 2020. Multiple logistic regression analyses showed that in the male population, the HU ratio (OR) of vitamin B1 intake in Q2 to Q5 compared with the lowest quintile (Q1) was 0.75 (95% CI 0.52, 1.09), 0.70 (95% CI 0.48, 1.02), 0.66 (95% CI 0.44, 0.99) and 0.55 (95% CI 0.34, 0.90). The P for trend was 0.028. In women, the ORs for vitamin B1 intake Q2 to Q5 were 0.87 (95% CI 0.64, 1.19), 0.97 (0.68-1.38), 1.05 (0.69-1.60) and 0.75 (0.42-1.34), respectively. The P for trend was 0.876. The RCS curve revealed a linear relationship between vitamin B1 intake and the risk of hyperuricemia in men (P nonlinear = 0.401). Smoothed curve fitting demonstrated a negative association between vitamin B1 intake and serum uric acid concentration in men, whereas there was no significant association between dietary vitamin B1 intake and the risk of hyperuricemia in women. In the US adult population, dietary vitamin B1 intake was negatively associated with hyperuricemia in males.


Subject(s)
Hyperuricemia , Nutrition Surveys , Thiamine , Uric Acid , Humans , Hyperuricemia/epidemiology , Hyperuricemia/blood , Hyperuricemia/etiology , Male , Female , Middle Aged , Adult , Cross-Sectional Studies , Uric Acid/blood , Thiamine/administration & dosage , Thiamine/blood , Prevalence , Diet , Odds Ratio , Risk Factors , Aged , United States/epidemiology
14.
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
15.
Front Endocrinol (Lausanne) ; 15: 1378645, 2024.
Article in English | MEDLINE | ID: mdl-39027467

ABSTRACT

Objective: Hyperuricaemia and gout are common metabolic disorders. However, the causal relationships between blood metabolites and serum urate levels, as well as gout, remain unclear. A systematic evaluation of the causal connections between blood metabolites, hyperuricemia, and gout could enhance early screening and prevention of hyperuricemia and gout in clinical settings, providing novel insights and approaches for clinical treatment. Methods: In this study, we employed a bidirectional two-sample Mendelian randomization analysis utilizing data from a genome-wide association study involving 7,286 participants, encompassing 486 blood metabolites. Serum urate and gout data were sourced from the Chronic Kidney Disease Genetics consortium, including 288,649 participants for serum urate and 9,819 African American and 753,994 European individuals for gout. Initially, LDSC methodology was applied to identify blood metabolites with a genetic relationship to serum urate and gout. Subsequently, inverse-variance weighting was employed as the primary analysis method, with a series of sensitivity and pleiotropy analyses conducted to assess the robustness of the results. Results: Following LDSC, 133 blood metabolites exhibited a potential genetic relationship with serum urate and gout. In the primary Mendelian randomization analysis using inverse-variance weighting, 19 blood metabolites were recognized as potentially influencing serum urate levels and gout. Subsequently, the IVW p-values of potential metabolites were corrected using the false discovery rate method. We find leucine (IVW P FDR = 0.00004), N-acetylornithine (IVW P FDR = 0.0295), N1-methyl-3-pyridone-4-carboxamide (IVW P FDR = 0.0295), and succinyl carnitine (IVW P FDR = 0.00004) were identified as significant risk factors for elevated serum urate levels. Additionally, 1-oleoylglycerol (IVW P FDR = 0.0007) may lead to a substantial increase in the risk of gout. Succinyl carnitine exhibited acceptable weak heterogeneity, and the results for other blood metabolites remained robust after sensitivity, heterogeneity, and pleiotropy testing. We conducted an enrichment analysis on potential blood metabolites, followed by a metabolic pathway analysis revealing four pathways associated with serum urate levels. Conclusion: The identified causal relationships between these metabolites and serum urate and gout offer a novel perspective, providing new mechanistic insights into serum urate levels and gout.


Subject(s)
Genome-Wide Association Study , Gout , Hyperuricemia , Mendelian Randomization Analysis , Metabolic Networks and Pathways , Uric Acid , Humans , Gout/genetics , Gout/blood , Gout/epidemiology , Uric Acid/blood , Metabolic Networks and Pathways/genetics , Hyperuricemia/blood , Hyperuricemia/genetics , Hyperuricemia/epidemiology , Polymorphism, Single Nucleotide , Female , Male
16.
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
17.
Nutrients ; 16(14)2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39064767

ABSTRACT

Although patients with hyperuricemia and gout often have dyslipidemia, the effects of febuxostat, a xanthine oxidase inhibitor, on their lipid profiles are unclear. Thus, we performed a sub-analysis of the randomized PRIZE study in which the effects of febuxostat on carotid atherosclerosis were investigated in patients with hyperuricemia. The participants were randomized to the febuxostat or control group. The primary endpoint of this sub-analysis was changes in the patients' non-high-density lipoprotein cholesterol (HDL-C) levels from baseline to 6-month follow-up. Correlations between the changes in lipid profiles and cardiometabolic parameters were also evaluated. In total, 456 patients were included. From baseline to 6 months, non-HDL-C levels were significantly reduced in the febuxostat group (-5.9 mg/dL, 95% confidence interval [CI]: -9.1 to -2.8 mg/dL, p < 0.001), but not in the control group (-1.3 mg/dL, 95% CI: -4.4 to 1.8, p = 0.348). The reduction in non-HDL-C levels was more pronounced in women and correlated with changes in serum uric acid and estimated glomerular filtration rate levels only in the febuxostat group. In patients with hyperuricemia, febuxostat treatment was associated with reduced non-HDL-C levels from baseline to the 6-month follow-up compared to the control treatment, suggesting that the lipid-lowering effect of febuxostat should be considered when targeting dyslipidemia.


Subject(s)
Febuxostat , Hyperuricemia , Lipids , Xanthine Oxidase , Humans , Febuxostat/therapeutic use , Febuxostat/pharmacology , Hyperuricemia/drug therapy , Hyperuricemia/blood , Xanthine Oxidase/antagonists & inhibitors , Male , Female , Middle Aged , Lipids/blood , Aged , Uric Acid/blood , Gout Suppressants/therapeutic use , Gout Suppressants/pharmacology , Cholesterol, HDL/blood , Carotid Artery Diseases/drug therapy , Carotid Artery Diseases/blood , Dyslipidemias/drug therapy , Dyslipidemias/blood , Glomerular Filtration Rate/drug effects
18.
Environ Sci Technol ; 58(29): 12875-12887, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38980177

ABSTRACT

There has been widespread concern about the health hazards of per- and polyfluoroalkyl substances (PFAS), which may be the risk factor for hyperuricemia with evidence still insufficient in the general population in China. Here, we conducted a nationwide study involving 9,580 adults aged 18 years or older from 2017 to 2018, measured serum concentrations of uric acid and PFAS (PFOA, PFOS, 6:2 Cl-PFESA, PFNA, PFHxS) in participants, to assess the associations of individual PFAS with hyperuricemia, and estimated a joint effect of PFAS mixtures. We found positive associations of higher serum PFAS with elevated odds of hyperuricemia in Chinese adults, with the greatest contribution from PFOA (69.37%). The nonmonotonic dose-response (NMDR) relationships were observed for 6:2 Cl-PFESA and PFHxS with hyperuricemia. Participants with less marine fish consumption, overweight, and obesity may be the sensitive groups to the effects of PFAS on hyperuricemia. We highlight the potential health hazards of legacy long-chain PFAS (PFOA) once again because of the higher weights of joint effects. This study also provides more evidence about the NMDR relationships in PFAS with hyperuricemia and emphasizes a theoretical basis for public health planning to reduce the health hazards of PFAS in sensitive groups.


Subject(s)
Hyperuricemia , Hyperuricemia/epidemiology , Hyperuricemia/blood , Humans , Cross-Sectional Studies , Adult , Male , Female , Fluorocarbons/blood , Middle Aged , China/epidemiology , Uric Acid/blood
19.
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
20.
Nutr Metab Cardiovasc Dis ; 34(9): 2203-2216, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39003131

ABSTRACT

BACKGROUND AND AIMS: The objective of this research was to explore the associations between dietary PUFAs intake and hyperuricemia risk. METHODS AND RESULTS: Based on the National Health and Nutrition Examination Survey (NHANES) 2003-2015, all eligible individuals were divided into hyperuricemia and non-hyperuricemia groups based on diagnostic criteria for hyperuricemia (serum uric acid >420 µmol/L for men and >360 µmol/L for women). Multivariate-adjusted logistic regression was employed to explore the relationship between dietary PUFAs intake and hyperuricemia risk. Total PUFAs and their subtypes were modeled to isocalorically replace saturated fatty acids (SFAs) and monounsaturated fatty acids (MUFAs). Higher intake of n-3 PUFAs, n-6 PUFAs, linoleic acid (LA), alpha-linoleic acid (ALA), and non-marine PUFAs intake correlated with decreased hyperuricemia risk, with adjusted odds ratio (OR) and 95% confidence interval (95%CIs) were 0.77 (0.63, 0.93), 0.75 (0.61, 0.92), 0.75 (0.61, 0.91), 0.69 (0.55, 0.87), and 0.73 (0.59, 0.91), respectively. Replacing 5% of total energy intake from SFAs with isocaloric PUFAs was associated with decreased odds of hyperuricemia in men (0.69 (0.57, 0.84)) and in individuals (0.81 (0.71, 0.92)). Similar trends were observed in the substitution of SFAs with non-marine PUFAs in men (0.87 (0.80, 0.94)) and in all individuals (0.92 (0.88, 0.98)). Sensitivity analyses exhibited consistent results with primary analyses. CONCLUSION: Higher dietary intake of n-3 PUFAs, n-6 PUFAs, LA, ALA, and non-marine PUFAs was associated with decreased hyperuricemia risk. These results support the recommendation to substitute SFAs with PUFAs in diet.


Subject(s)
Biomarkers , Hyperuricemia , Nutrition Surveys , Protective Factors , Uric Acid , Humans , Hyperuricemia/epidemiology , Hyperuricemia/diagnosis , Hyperuricemia/blood , Hyperuricemia/prevention & control , Male , Female , Middle Aged , Uric Acid/blood , Adult , Risk Factors , Risk Assessment , Cross-Sectional Studies , Biomarkers/blood , United States/epidemiology , Recommended Dietary Allowances , Aged , Time Factors , Fatty Acids, Omega-6/administration & dosage , Fatty Acids, Unsaturated/administration & dosage , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL