1
|
Zhang X, He Z, Wang S, Zhang S, Song D. A pure near-infrared platform with dual-readout capability employing upconversion fluorescence and colorimetry for biosensing of uric acid. Talanta 2025; 291:127900. [PMID: 40056650 DOI: 10.1016/j.talanta.2025.127900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/25/2025] [Accepted: 03/04/2025] [Indexed: 03/10/2025]
Abstract
Exploring an accurate uric acid (UA) detection method is of paramount importance for early disease diagnosis. In this study, we have developed a novel near-infrared (NIR) probe that integrates upconversion nanoparticles (UCNPs) with polymetallic oxomolybdate (POM) clusters to achieve precise UA quantification. The strong absorption of POM peaking at 825 nm effectively quenched the fluorescence emission of UCNPs at 806 nm under 980 nm laser excitation through the resonance energy transfer effect. Upon introducing UA along with uricase, hydrogen peroxide generated from the catalytic reaction significantly diminished POM absorption, thereby restoring UCNP fluorescence by up to 19.5-fold. By leveraging the distinctive features of NIR dual-readout and NIR excitation, the interference from biological samples can be significantly mitigated. Consequently, the probe demonstrated excellent selectivity and sensitivity towards UA. For the colorimetric assay, the linear range for UA detection was 5-100 μM with a low detection limit of 0.283 μM, while the fluorescence method demonstrated a linear range of 1-60 μM with a detection limit as low as 11.74 nM. We successfully and accurately quantified UA in human serum, highlighting its great potential for biochemical and clinical applications.
Collapse
Affiliation(s)
- Xinglong Zhang
- College of Chemistry and Chemical Engineering, Yulin University, Yulin, 719000, PR China
| | - Zuming He
- College of Chemistry and Chemical Engineering, Yulin University, Yulin, 719000, PR China
| | - Shuyan Wang
- College of Chemistry and Chemical Engineering, Yulin University, Yulin, 719000, PR China
| | - Shuai Zhang
- Xingyuan Hospital of Yulin, Yulin, 719000, PR China
| | - Dan Song
- College of Chemistry and Chemical Engineering, Yulin University, Yulin, 719000, PR China.
| |
Collapse
|
2
|
Dempsey B, Pereira da Silva B, Cruz LC, Vileigas D, Silva ARM, Pereira da Silva R, Meotti FC. Unraveling the effects of uric acid on endothelial cells: A global proteomic study. Redox Biol 2025; 82:103625. [PMID: 40203480 PMCID: PMC12005352 DOI: 10.1016/j.redox.2025.103625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/27/2025] [Accepted: 03/29/2025] [Indexed: 04/11/2025] Open
Abstract
This work aims to understand how normouricemic levels of uric acid can induce endothelial dysfunction seeking global proteomic alterations in Human Umbilical Vein cells (HUVEC). It reveals significant alterations in redox-sensitive and antioxidant proteins, chaperones, and proteins associated with cell migration and adhesion in response to uric acid exposure. Monitoring cellular oxidation with the roGFP2-Grx1 probe proved increased oxidation levels induced by uric acid, which can be attenuated by peroxidasin (PXDN) inhibition, suggesting a regulatory role for PXDN in mitigating oxidative stress induced by uric acid. As a consequence of uric acid oxidation and the formation of reactive intermediate, we identified adducts in proteins (+140 kDa) in a novel post-translation modification named uratylation. Increased misfolded protein levels and p62 aggregation were also found, indicating disturbances in cellular proteostasis. Furthermore, uric acid promoted monocyte adhesion and upregulated ICAM and VCAM protein levels, implicating a pro-inflammatory response in endothelial cells. These findings provide critical insights into the molecular mechanisms underlying vascular damage associated with uric acid.
Collapse
Affiliation(s)
- Bianca Dempsey
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | | | - Litiele Cezar Cruz
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Danielle Vileigas
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Amanda R M Silva
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | | | - Flavia Carla Meotti
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
3
|
Liu C, Mao Q, Zhang B, Fu X, Zhang T, Wang S. A patent review of xanthine oxidase inhibitors (2021-present). Expert Opin Ther Pat 2025; 35:79-89. [PMID: 39731464 DOI: 10.1080/13543776.2024.2446222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/24/2024] [Accepted: 12/17/2024] [Indexed: 12/29/2024]
Abstract
INTRODUCTION Xanthine oxidase (XO) catalyzes the oxidation of both hypoxanthine and xanthine in the last two steps of the purine metabolic pathway, serving as a rate-limiting enzyme for uric acid production as well as a key target for the treatment of gout and other hyperuricemia-related conditions. AREAS COVERED This paper reviews XO inhibitors in patents from 2021 to the present. We summarize in detail the structural classes and characteristics, in vitro and in vivo biological results, and structure‒activity relationships of synthetic inhibitors, as well as the sources, specific structures, research methods, and biological activities of XO inhibitors from natural products. EXPERT OPINION (1) Benefiting from the discovery of many high-affinity inhibitors, the binding modes of small molecules in the active pocket of XO have been further elucidated, and this information will contribute to future development; (2) natural products remain one of the important sources in the discovery of XO inhibitors; (3) with a deeper exploration of XO and URAT1 targets, XO/URAT1 dual target inhibitors may be a future research hotspot; and (4) the search for high-affinity, small-molecule scaffolds remains a key challenge and an important direction for the future development.
Collapse
Affiliation(s)
- Chang Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Qing Mao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Bing Zhang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Xuefeng Fu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Tingjian Zhang
- School of Pharmacy, China Medical University, Shenyang, China
| | - Shaojie Wang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
4
|
Zhou B, Fu J, Yuan Y, Han F, Huo K, Chu PK, Zhang X. Potential-dependent simultaneous detection of uric acid and glucose using dual-function Ni@CNT supported carbon fiber electrodes. Microchem J 2024; 205:111244. [DOI: 10.1016/j.microc.2024.111244] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
5
|
Li KW, Raza F, Jiang LD, Su J, Qiu MF. Clerodendranthus Spicatus: A review of its active compounds, mechanisms of action, and clinical studies in urinary diseases. Fitoterapia 2024; 177:106082. [PMID: 38901804 DOI: 10.1016/j.fitote.2024.106082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/09/2024] [Accepted: 06/16/2024] [Indexed: 06/22/2024]
Abstract
Clerodendranthus spicatus (Thunb.) C.Y.Wu (CS) is a widely studied plant that shows potential in treating urinary diseases. Previous studies have focused on its chemical composition, pharmacological effects, and clinical applications. This review aims to provide a comprehensive summary and evaluation of the existing literature on CS. It also suggests future research directions to increase our understanding of its medicinal value. 129 pieces of literature were selected from several databases, including PubMed, Web of Science, China National Knowledge Infrastructure (CNKI), Wan-fang Database, and Google Scholar, and were analyzed. Forty-five active compounds of CS have pharmacological effects such as lowering uric acid, anti-inflammation, anti-oxidation, and kidney protection. The potential mechanisms of these effects may be related to inhibiting transforming growth factor β1 (TGF-β1) activation, reducing inflammatory factors such as IL-8, IL-1β, TNF-α, PGE2, IFN-γ, and IL-6 levels, suppressing the activation of NF-κB, JAK/STAT pathway, enhancing the clearance of ROS, MDA DPPH·, and O2 ̇ -, and regulating the expression of apoptosis-related pathways and proteins. This paper also discusses the quality control of CS and its efficacy and safety in treating urinary diseases. The study concludes that CS has a high potential for treating urinary diseases. Future studies should focus on observing the metabolic changes of CS active compounds in vivo and investigating the effects of CS on key signaling pathways. Additionally, more standardized and reasonable clinical studies and safety evaluation experiments should be conducted to obtain more clinical data.
Collapse
Affiliation(s)
- Kun-Wei Li
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Liang-di Jiang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ming-Feng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
6
|
Cai N, Chen M, Feng P, Zheng Q, Zhu X, Yang S, Zhang Z, Wang Y. Relationships between obesity and prevalence of gout in patients with type 2 diabetes mellitus: a cross-sectional population-based study. BMC Endocr Disord 2024; 24:137. [PMID: 39090627 PMCID: PMC11295670 DOI: 10.1186/s12902-024-01672-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND The purpose of this study was to investigate the relationships between generalized, abdominal, and visceral fat obesity and the prevalence of gout in patients with type 2 diabetes mellitus (T2DM). METHODS Data were obtained from the electronic medical databases of the National Metabolic Management Center (MMC) of Yuhuan Second People's Hospital and Taizhou Central Hospital (Taizhou University Hospital) between September 2017 and June 2023. Four obesity indicators were analyzed: waist circumference (WC), waist-to-hip ratio (WHR), body mass index (BMI), and visceral fat area (VFA). The relationships between these parameters and gout prevalence were analyzed using multivariate logistic regression and restricted cubic spline (RCS) analyses. Receiver operating characteristic (ROC) curves were used to evaluate the diagnostic efficacy of the four parameters for gout. RESULTS This cross-sectional study enrolled 10,535 participants (600 cases and 9,935 controls). Obesity was more common in patients with gout, and the obesity indicators were markedly higher in this group. After adjustment for confounders, obesity, as defined by BMI, WC, WHR, and VFA, was found to be associated with greater gout prevalence, with odds ratios (OR) of 1.775, 1.691, 1.858, and 1.578, respectively (P < 0.001). The gout odds ratios increased markedly in relation to the obesity indicator quartiles (P-value for trend < 0.001), and the obesity indicators were positively correlated with gout prevalence, as shown using RCS. The area under the ROC curve values for BMI, WC, WHR, and VFA were 0.629, 0.651, 0.634, and 0.633, respectively. CONCLUSION Obesity-whether general, abdominal, or visceral fat obesity-was positively linked with elevated gout risk. But uncovering the causality behind the relationship requires further prospective study. Obesity indicators (BMI, WC, WHR, and VFA) may have potential value for diagnosing gout in clinical practice.
Collapse
Affiliation(s)
- Ningyu Cai
- Department of Orthopedics, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Mengdie Chen
- Department of Endocrinology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Ping Feng
- Department of Endocrinology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Qidong Zheng
- Department of Internal Medicine, Yuhuan Second People's Hospital, Yuhuan, China
| | - Xianping Zhu
- Department of Orthopedics, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Suqing Yang
- Customer Service Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Zhaobo Zhang
- Department of Orthopedics, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Yiyun Wang
- Department of Internal Medicine, Yuhuan Second People's Hospital, Yuhuan, China.
| |
Collapse
|
7
|
Zhang X, Zhang X, Li X, Zhao X, Wei G, Shi J, Yang Y, Fan S, Zhao J, Zhu K, Du J, Guo J, Cao W. Association between serum uric acid levels and diabetic peripheral neuropathy in type 2 diabetes: a systematic review and meta-analysis. Front Endocrinol (Lausanne) 2024; 15:1416311. [PMID: 39072278 PMCID: PMC11272597 DOI: 10.3389/fendo.2024.1416311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/25/2024] [Indexed: 07/30/2024] Open
Abstract
Background The evidence supporting a connection between elevated serum uric acid (SUA) levels and diabetic peripheral neuropathy (DPN) is controversial. The present study performed a comprehensive evaluation of this correlation by conducting a systematic review and meta-analysis of relevant research. Method PubMed, Web of Science (WOS), Embase, and the Cochrane Library were searched for published literature from the establishment of each database to January 8, 2024. In total, 5 cohort studies and 15 cross-sectional studies were included, and 2 researchers independently screened and extracted relevant data. R 4.3.0 was used to evaluate the included literature. The present meta-analysis evaluated the relationship between SUA levels and the risk of DPN in type 2 diabetes (T2DM) by calculating the ratio of means (RoM) and 95% confidence intervals (CIs) using the method reported by JO Friedrich, and it also analyzed continuous outcome measures using standardized mean differences (SMDs) and 95% CIs to compare SUA levels between DPN and non-DPN groups. Funnel plot and Egger's test were used to assess publication bias. Sensitivity analysis was conducted by sequentially removing each study one-by-one. Results The meta-analysis included 20 studies, with 12,952 T2DM patients with DPN and 16,246 T2DM patients without DPN. There was a significant correlation between SUA levels and the risk of developing DPN [odds ratio (OR) = 1.23; 95% CI: 1.07-1.41; p = 0.001]. Additionally, individuals with DPN had higher levels of SUA compared to those without DPN (SMD = 0.4; 95% CI: -0.11-0.91; p < 0.01). Conclusion T2DM patients with DPN have significantly elevated SUA levels, which correlate with a heightened risk of peripheral neuropathy. Hyperuricemia (HUA) may be a risk indicator for assessing the risk of developing DPN in T2DM patients. Systematic review registration https://www.crd.york.ac.uk/PROSPERO, identifier CRD42024500373.
Collapse
Affiliation(s)
- Xieyu Zhang
- Department of Rheumatology, Wangjing Hospital, China Academy of Chinese Medicine Science, Beijing, China
| | - Xinwen Zhang
- Department of Rheumatology, Wangjing Hospital, China Academy of Chinese Medicine Science, Beijing, China
| | - Xiaoxu Li
- Department of Rheumatology, Wangjing Hospital, China Academy of Chinese Medicine Science, Beijing, China
| | - Xin Zhao
- Department of Rheumatology, Wangjing Hospital, China Academy of Chinese Medicine Science, Beijing, China
| | - Guangcheng Wei
- Department of Rheumatology, Wangjing Hospital, China Academy of Chinese Medicine Science, Beijing, China
| | - Jinjie Shi
- Department of Rheumatology, Wangjing Hospital, China Academy of Chinese Medicine Science, Beijing, China
| | - Yue Yang
- Department of Rheumatology, Wangjing Hospital, China Academy of Chinese Medicine Science, Beijing, China
| | - Su Fan
- Department of Rheumatology, Wangjing Hospital, China Academy of Chinese Medicine Science, Beijing, China
| | - Jiahe Zhao
- Department of Rheumatology, Wangjing Hospital, China Academy of Chinese Medicine Science, Beijing, China
| | - Ke Zhu
- Department of Rheumatology, Wangjing Hospital, China Academy of Chinese Medicine Science, Beijing, China
| | - Jieyang Du
- Department of Rheumatology, Wangjing Hospital, China Academy of Chinese Medicine Science, Beijing, China
| | - Junyi Guo
- Robotics Movement Department, Amazon, Boston, MA, United States
| | - Wei Cao
- Department of Rheumatology, Wangjing Hospital, China Academy of Chinese Medicine Science, Beijing, China
| |
Collapse
|
8
|
Wang K, Wu J, Deng M, Nie J, Tao F, Li Q, Luo X, Xia F. Associations of oxidative balance score with hyperuricemia and gout among American adults: a population-based study. Front Endocrinol (Lausanne) 2024; 15:1354704. [PMID: 38988995 PMCID: PMC11233537 DOI: 10.3389/fendo.2024.1354704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 05/31/2024] [Indexed: 07/12/2024] Open
Abstract
Objective The current study aimed to assess the relationships between oxidative balance score (OBS) and OBS subclasses (dietary and lifestyle OBS) with risks of hyperuricemia (HUA) and gout among American adults. Methods Participants in the National Health and Nutrition Examination Survey from 2007 to 2018 were initially recruited and then the final sample was restricted to adults without missing values about serum uric acid, gout, OBS, and covariates. Rao-Scott adjusted chi-square test and analysis of variance were utilized to compare the baseline characteristics in adults of different quartiles of OBS, while the weighted stepped logistic regression models were used to explore the associations of overall, dietary, and lifestyle OBS with the risks of HUA and gout. Weighted restricted cubic spline analyses were conducted to explore the nonlinear dose-response associations. Results The final sample consisted of 22,705 participants aged 20 years and older, which was representative of approximately 197.3 million non-institutionalized American adults. HUA and gout prevalence decreased with OBS quartiles. Compared with adults in the first quartile of OBS, those in the second (OR: 0.85, 95% CI: 0.72-0.99), third (OR: 0.71, 95% CI: 0.58-0.85), and fourth (OR: 0.48, 95% CI: 0.38-0.61) quartiles of OBS had reduced risks of hyperuricemia. Similarly, adults in the second (OR: 0.70, 95% CI: 0.51-0.97) quartile of OBS was associated with lower gout risk in comparison to adults in the lowest quartile. Regarding OBS subclasses, dietary and lifestyle OBS were both negatively correlated with the risk of HUA, and only higher lifestyle OBS was significantly associated with lower gout risk. Furthermore, the subgroup analyses and interaction effects also substantiated similar effects. Significant nonlinear dose-response relationships were observed between overall, dietary, and lifestyle OBS with HUA risk as well as that of lifestyle OBS with gout risk. Conclusion This study strongly suggests the significant negative associations of OBS with HUA and gout in American adults and provides a dietary and lifestyle guideline to reduce the risks.
Collapse
Affiliation(s)
- Kai Wang
- Department of Public Health, Wuhan Fourth Hospital, Wuhan, China
| | - Jinyi Wu
- Department of Public Health, Wuhan Fourth Hospital, Wuhan, China
| | - Minggang Deng
- Department of Psychiatry, Wuhan Mental Health Center, Wuhan, China
- Department of Psychiatry, Wuhan Hospital for Psychotherapy, Wuhan, China
| | - Jiaqi Nie
- XiaoGan Center for Disease Control and Prevention, Xiaogan, China
| | - Fengxi Tao
- Department of Public Health, Wuhan Fourth Hospital, Wuhan, China
| | - Qingwen Li
- Department of Public Health, Wuhan Fourth Hospital, Wuhan, China
| | - Xin Luo
- Department of Public Health, Wuhan Fourth Hospital, Wuhan, China
| | - Fang Xia
- Department of Public Health, Wuhan Fourth Hospital, Wuhan, China
| |
Collapse
|
9
|
Huang D, Li W, Zhao Y, Xie C, Luo X, Wu F, Xu Z, Sun Q, Liu G. Design, synthesis, and biological evaluation of 5-(1H-indol-5-yl)isoxazole-3-carboxylic acids as novel xanthine oxidase inhibitors. Eur J Med Chem 2024; 271:116443. [PMID: 38691887 DOI: 10.1016/j.ejmech.2024.116443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 04/07/2024] [Accepted: 04/19/2024] [Indexed: 05/03/2024]
Abstract
Xanthine oxidase (XO) is a key enzyme for the production of uric acid in the human body. XO inhibitors (XOIs) are clinically used for the treatment of hyperuricemia and gout, as they can effectively inhibit the production of uric acid. Previous studies indicated that both indole and isoxazole derivatives have good inhibitory effects against XO. Here, we designed and synthesized a novel series of N-5-(1H-indol-5-yl)isoxazole-3-carboxylic acids according to bioisosteric replacement and hybridization strategies. Among the obtained target compounds, compound 6c showed the best inhibitory activity against XO with an IC50 value of 0.13 μM, which was 22-fold higher than that of the classical antigout drug allopurinol (IC50 = 2.93 μM). Structure-activity relationship analysis indicated that the hydrophobic group on the nitrogen atom of the indole ring is essential for the inhibitory potencies of target compounds against XO. Enzyme kinetic studies proved that compound 6c acted as a mixed-type XOI. Molecular docking studies showed that the target compound 6c could not only retain the key interactions similar to febuxostat at the XO binding site but also generate some new interactions, such as two hydrogen bonds between the oxygen atom of the isoxazole ring and the amino acid residues Ser876 and Thr1010. These results indicated that 5-(1H-indol-5-yl)isoxazole-3-carboxylic acid might be an efficacious scaffold for designing novel XOIs and compound 6c has the potential to be used as a lead for further the development of novel anti-gout candidates.
Collapse
Affiliation(s)
- Dongqian Huang
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China
| | - Wenye Li
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China
| | - Yilan Zhao
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China
| | - Cheng Xie
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China
| | - Xiaogang Luo
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China; School of Materials Science and Engineering, Zhengzhou University, No.100 Science Avenue, Zhengzhou City, 450001, Henan Province, PR China
| | - Fengshou Wu
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China
| | - Zhiqiang Xu
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China
| | - Qi Sun
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China.
| | - Genyan Liu
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China.
| |
Collapse
|
10
|
Li Y, Shi Z, Wu X, Miao W, Yang Z, Lou X, Sun W, Guo C, Li CM. Graphdiyne marries PEDOT:PSS to form high-stable heterostructure from 2-unstable components toward ultra-low detection limit of uric acid detection in sweat. Mikrochim Acta 2024; 191:243. [PMID: 38575711 DOI: 10.1007/s00604-024-06311-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 03/11/2024] [Indexed: 04/06/2024]
Abstract
PEDOT PSS has been used as a biomimetic uric acid (UA) sensor but suffers from unfortunate low detection limit (LOD), narrow detection range and poor stability. Herein, we get graphdiyne (GDY) marry PEDOT:PSS to create a very stable GDY@PEDOT:PSS heterostructure for a biomimetic UA sensor, which accomplishes the lowest LOD (6 nM), the widest detection range (0.03 μM-7 mM) and the longest stability (98.1% for 35 days) among the related UA sensors. The sensor was successfully used to in situ real-time detection of UA in sweat. The enhancement mechanisms of the sensor were investigated, and results discover that C≡C of GDY and C = C of PEDOT:PSS can cross-link each other by π-π interactions, making not only the former strongly resistant against oxidation deterioration, but also causes the latter to efficiently prevent water swelling of polymer for poor conductivity, thereby leading to high stability from both components. While the stabilized heterostructure can also offer more active sites by enhanced absorption of UA via π-π interactions for highly sensitive detection of UA. This work holds great promise for a practical sweat UA sensor while providing scientific insight to design a stable and electrocatalytically active structure from two unstable components.
Collapse
Affiliation(s)
- Yunpeng Li
- Institute of Materials Science and Devices, School of Materials Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215011, China
| | - Zhuanzhuan Shi
- Institute of Materials Science and Devices, School of Materials Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215011, China.
| | - Xiaoshuai Wu
- Institute of Materials Science and Devices, School of Materials Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215011, China
| | - Wenting Miao
- Institute of Materials Science and Devices, School of Materials Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215011, China
| | - Zhengyi Yang
- Institute of Materials Science and Devices, School of Materials Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215011, China
| | - Xinyu Lou
- Institute of Materials Science and Devices, School of Materials Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215011, China
| | - Wei Sun
- College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, China
| | - Chunxian Guo
- Institute of Materials Science and Devices, School of Materials Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215011, China.
| | - Chang Ming Li
- Institute of Materials Science and Devices, School of Materials Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215011, China.
| |
Collapse
|
11
|
Yang Z, Guo J, Wang L, Zhang J, Ding L, Liu H, Yu X. Nanozyme-Enhanced Electrochemical Biosensors: Mechanisms and Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307815. [PMID: 37985947 DOI: 10.1002/smll.202307815] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/22/2023] [Indexed: 11/22/2023]
Abstract
Nanozymes, as innovative materials, have demonstrated remarkable potential in the field of electrochemical biosensors. This article provides an overview of the mechanisms and extensive practical applications of nanozymes in electrochemical biosensors. First, the definition and characteristics of nanozymes are introduced, emphasizing their significant role in constructing efficient sensors. Subsequently, several common categories of nanozyme materials are delved into, including metal-based, carbon-based, metal-organic framework, and layered double hydroxide nanostructures, discussing their applications in electrochemical biosensors. Regarding their mechanisms, two key roles of nanozymes are particularly focused in electrochemical biosensors: selective enhancement and signal amplification, which crucially support the enhancement of sensor performance. In terms of practical applications, the widespread use of nanozyme-based electrochemical biosensors are showcased in various domains. From detecting biomolecules, pollutants, nucleic acids, proteins, to cells, providing robust means for high-sensitivity detection. Furthermore, insights into the future development of nanozyme-based electrochemical biosensors is provided, encompassing improvements and optimizations of nanozyme materials, innovative sensor design and integration, and the expansion of application fields through interdisciplinary collaboration. In conclusion, this article systematically presents the mechanisms and applications of nanozymes in electrochemical biosensors, offering valuable references and prospects for research and development in this field.
Collapse
Affiliation(s)
- Zhongwei Yang
- Institute for Advanced Interdisciplinary Research (iAIR), School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, P. R. China
| | - Jiawei Guo
- Institute for Advanced Interdisciplinary Research (iAIR), School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, P. R. China
| | - Longwei Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, University of Chinese Academy of Science, Beijing, 100190, P. R. China
| | - Jian Zhang
- Division of Systems and Synthetic Biology, Department of Life Sciences, Chalmers University of Technology, Göteborg, 41296, Sweden
| | - Longhua Ding
- Institute for Advanced Interdisciplinary Research (iAIR), School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, P. R. China
| | - Hong Liu
- Institute for Advanced Interdisciplinary Research (iAIR), School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, P. R. China
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, P. R. China
| | - Xin Yu
- Institute for Advanced Interdisciplinary Research (iAIR), School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, P. R. China
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Qingdao University of Science and Technology, Qingdao, 266042, P. R. China
| |
Collapse
|
12
|
Ma Q, Lim CS. Molecular Activation of NLRP3 Inflammasome by Particles and Crystals: A Continuing Challenge of Immunology and Toxicology. Annu Rev Pharmacol Toxicol 2024; 64:417-433. [PMID: 37708431 PMCID: PMC10842595 DOI: 10.1146/annurev-pharmtox-031023-125300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
Particles and crystals constitute a unique class of toxic agents that humans are constantly exposed to both endogenously and from the environment. Deposition of particulates in the body is associated with a range of diseases and toxicity. The mechanism by which particulates cause disease remains poorly understood due to the lack of mechanistic insights into particle-biological interactions. Recent research has revealed that many particles and crystals activate the NLRP3 inflammasome, an intracellular pattern-recognition receptor. Activated NLRP3 forms a supramolecular complex with an adaptor protein to activate caspase 1, which in turn activates IL-1β and IL-18 to instigate inflammation. Genetic ablation and pharmacological inhibition of the NLRP3 inflammasome dampen inflammatory responses to particulates. Nonetheless, how particulates activate NLRP3 remains a challenging question. From this perspective, we discuss our current understanding of and progress on revealing the function and mode of action of the NLRP3 inflammasome in mediating adaptive and pathologic responses to particulates in health and disease.
Collapse
Affiliation(s)
- Qiang Ma
- Receptor Biology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia, USA;
| | - Chol Seung Lim
- Receptor Biology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia, USA;
| |
Collapse
|
13
|
Fang ZE, Wang Y, Bian S, Qin S, Zhao H, Wen J, Liu T, Ren L, Li Q, Shi W, Zhao J, Yang H, Peng R, Wang Q, Bai Z, Xu G. Helenine blocks NLRP3 activation by disrupting the NEK7-NLRP3 interaction and ameliorates inflammatory diseases. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 122:155159. [PMID: 37931457 DOI: 10.1016/j.phymed.2023.155159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 09/19/2023] [Accepted: 10/19/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND The involvement of NLRP3 inflammasome is associated with the progress of numerous inflammatory conditions. However, there is currently no single compound used in the clinic. Search for the inhibitor of NLRP3 inflammasome from natural products is an attractive direction. The compound Helenin (Hel), which is obtained from Inula helenium L., is reported to have anti-inflammatory activities. However, the underlying molecular mechanisms and specific inflammatory signal pathway remains not well understood. PURPOSE This research aims to determine the impacts of Hel on NLRP3 inflammasome and the underlying mechanism involved, meanwhile also assessing its potential as a therapeutic intervention for inflammatory diseases mediated by NLRP3 overactivation. METHODS Pretreated with Hel in BMDMs (bone marrow-derived macrophages), then stimulated with NLRP3 triggers and measured the expression of active caspase-1 and interleukin 1β (IL-1β). Determination of intracellular K+ and Ca2+, ASC oligomerization and mitochondrial reactive oxygen species (mtROS) production were employed to explore the preliminary mechanism of Hel on NLRP3 activation. Subsequently, Co-immunoprecipitation was used to investigate protein-protein interaction and reduction of covalent bonds of Hel was to explore the binding mode between drugs and proteins. Finally, in vivo experiments, we utilized mouse lethal sepsis and monosodium urate(MSU)-induced peritonitis models to evaluate the effectiveness of Hel in inhibiting inflammatory diseases. RESULTS The findings revealed that Hel exhibited a specific blocking effect on NLRP3, with no impact on the assembly of NLRC4 and AIM2 inflammasome. Through the analysis of mechanisms targeting key upstream factors in NLRP3 activation, Hel inhibited NLRP3-dependent ASC oligomerization but did not regulating inflammasome priming, K+ efflux, Ca2+ influx, or mitochondrial damage and mtROS. Moreover, Hel effectively interrupted the binding of NEK7-NLRP3, which was dependent on the active double C=C of the α,β-unsaturated carbonyl units in Hel. In mouse models, Hel showed promising therapeutic effects in the treatment of NLRP3 overactivation-associated diseases, including the lethal sepsis and acute systemic inflammation induced by lipopolysaccharide (LPS) and peritonitis induced by MSU. CONCLUSION Our results indicate that Hel dependent α,β-unsaturated carbonyl units interrupt the formation of the NLRP3-NEK7 interaction, thereby blocks the inflammasome assemblage and activation. These fundings would suggest that Hel is a promising inhibitor for treating diseases driven by NLRP3 overactivation.
Collapse
Affiliation(s)
- Zhi-E Fang
- Department of Pharmacy, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400021, China; Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
| | - Yan Wang
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China; School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100105, China
| | - Shuyi Bian
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Shuanglin Qin
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China; School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Huanying Zhao
- Core Facilities Center, Capital Medical University, Beijing, 100069, China
| | - Jincai Wen
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
| | - Tingting Liu
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
| | - Lutong Ren
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
| | - Qiang Li
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
| | - Wei Shi
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
| | - Jia Zhao
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
| | - Huijie Yang
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
| | - Rui Peng
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Qin Wang
- Department of Pharmacy, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400021, China.
| | - Zhaofang Bai
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China.
| | - Guang Xu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
14
|
Frazaei MH, Nouri R, Arefnezhad R, Pour PM, Naseri M, Assar S. A Review of Medicinal Plants and Phytochemicals for the Management of Gout. Curr Rheumatol Rev 2024; 20:223-240. [PMID: 37828678 DOI: 10.2174/0115733971268037230920072503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/08/2023] [Accepted: 08/10/2023] [Indexed: 10/14/2023]
Abstract
Gout, characterized by elevated uric acid levels, is a common inflammatory joint disease associated with pain, joint swelling, and bone erosion. Existing treatments for gout often result in undesirable side effects, highlighting the need for new, safe, and cost-effective anti-gout drugs. Natural products, including medicinal plants and phytochemicals, have gained attention as potential sources of anti-gout compounds. In this review, we examined articles from 2000 to 2020 using PubMed and Google Scholar, focusing on the effectiveness of medicinal plants and phyto-chemicals in managing gout. Our findings identified 14 plants and nine phytochemicals with anti-gout properties. Notably, Teucrium polium, Prunus avium, Smilax riparia, Rhus coriaria, Foenic-ulum vulgare, Allium cepa, Camellia japonica, and Helianthus annuus exhibited the highest xa-thine oxidase inhibitory activity, attributed to their unique natural bioactive compounds such as phenolics, tannins, coumarins, terpenoids, and alkaloids. Herbal plants and their phytochemicals have demonstrated promising effects in reducing serum urate and inhibiting xanthine. This review aims to report recent studies on plants/phytochemicals derived from herbs beneficial in gout and their different mechanisms.
Collapse
Affiliation(s)
- Mohammad Hosein Frazaei
- Department of Pharmacology, Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Roghayeh Nouri
- Department of Pharmacology, Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Reza Arefnezhad
- Anatomical Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pardis Mohammadi Pour
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Naseri
- Department of Pharmacology, Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shirin Assar
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
15
|
Xie Y, Lin Z, Zhang J, Chen Y, Huang J, Tang H, Chen J, Lei Y, Qian Z. Virtual screening combined with experimental verification reveals the potential mechanism of Fuzitang decoction against Gouty Arthritis. Heliyon 2023; 9:e22650. [PMID: 38058447 PMCID: PMC10696199 DOI: 10.1016/j.heliyon.2023.e22650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 11/15/2023] [Accepted: 11/15/2023] [Indexed: 12/08/2023] Open
Abstract
Background and Purpose: Fuzitang decoction (FZT), a classic prescription of traditional Chinese medicine (TCM), has excellent efficacy in treating gouty arthritis (GA). However, the underlying molecular mechanism remains obscure. In the present study, we aimed to explore the underlying mechanisms of FZT in treating GA by virtual screening combined with experimental verification. Methods In this study, the active components of FZT and their corresponding targets were screened from the TCMSP database and TargetNet database. Then, the potential targets of FZT against GA were retrieved from multiple databases to generate a network. Protein-protein interaction, herbal-component-target, Gene Ontology (GO) enrichment, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were applied to identify potential targets and related signaling pathways. Furthermore, molecular docking simulation was applied to identify the interactions between the drug and targets. Finally, in vitro experiments were conducted to validate the potential targets and signaling pathways. Results In the present study, several crucial components, including kaempferol, luteolin, catechin, deoxyandrographolide, and perlolyrine in FZT, were obtained through network pharmacology, and several potential targets to treat GA were developed, such as PPARG, CYP3A4, PTGS2 (known as COX2), VEGFA, and CYP1A1. Experimental validation suggested that deoxyandrographolide significantly suppressed the expression of IL-1β, COX2, NLRP3 and IL-6 in inflammatory monocyte cells. Conclusions Our results identified a novel anti-inflammatory compound, deoxyandrographolide, which helps to explain the potential mechanism of FZT in treating GA and provides evidence to support FZT's clinical use.
Collapse
Affiliation(s)
- Yufeng Xie
- The Sixth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, 518000, China
| | - Zhongxiao Lin
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510000, China
| | - Jianmei Zhang
- The Sixth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Yun Chen
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, 518000, China
| | - Jianhao Huang
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, 518000, China
| | - Hong Tang
- The Sixth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Jieting Chen
- The Sixth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Yuhe Lei
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, 518000, China
| | - Ziliang Qian
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, 518000, China
| |
Collapse
|
16
|
Solangi AG, Tahira A, Waryani B, Chang AS, Pirzada T, Nafady A, Dawi EA, Saleem LMA, Padervand M, Haj Ismail AAK, Lv K, Vigolo B, Ibupoto ZH. Green-Mediated Synthesis of NiCo 2O 4 Nanostructures Using Radish White Peel Extract for the Sensitive and Selective Enzyme-Free Detection of Uric Acid. BIOSENSORS 2023; 13:780. [PMID: 37622866 PMCID: PMC10452471 DOI: 10.3390/bios13080780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/25/2023] [Accepted: 07/30/2023] [Indexed: 08/26/2023]
Abstract
The ability to measure uric acid (UA) non-enzymatically in human blood has been demonstrated through the use of a simple and efficient electrochemical method. A phytochemical extract from radish white peel extract improved the electrocatalytic performance of nickel-cobalt bimetallic oxide (NiCo2O4) during a hydrothermal process through abundant surface holes of oxides, an alteration of morphology, an excellent crystal quality, and increased Co(III) and Ni(II) chemical states. The surface structure, morphology, crystalline quality, and chemical composition were determined using a variety of analytical techniques, including powder X-ray diffraction (XRD), scanning electron microscopy (SEM), high-resolution transmission electron microscopy (HR-TEM), and X-ray photoelectron spectroscopy (XPS). The electrochemical characterization by CV revealed a linear range of UA from 0.1 mM to 8 mM, with a detection limit of 0.005 mM and a limit of quantification (LOQ) of 0.008 mM. A study of the sensitivity of NiCo2O4 nanostructures modified on the surface to UA detection with amperometry has revealed a linear range from 0.1 mM to 4 mM for detection. High stability, repeatability, and selectivity were associated with the enhanced electrochemical performance of non-enzymatic UA sensing. A significant contribution to the full outperforming sensing characterization can be attributed to the tailoring of surface properties of NiCo2O4 nanostructures. EIS analysis revealed a low charge-transfer resistance of 114,970 Ohms that offered NiCo2O4 nanostructures prepared with 5 mL of radish white peel extract, confirming an enhanced performance of the presented non-enzymatic UA sensor. As well as testing the practicality of the UA sensor, blood samples from human beings were also tested for UA. Due to its high sensitivity, stability, selectivity, repeatability, and simplicity, the developed non-enzymatic UA sensor is ideal for monitoring UA for a wide range of concentrations in biological matrixes.
Collapse
Affiliation(s)
- Abdul Ghaffar Solangi
- Institute of Chemistry, Shah Abdul Latif University Khairpur Mirs, Khairpur Mirs 66111, Pakistan; (A.G.S.); (A.T.); (T.P.)
| | - Aneela Tahira
- Institute of Chemistry, Shah Abdul Latif University Khairpur Mirs, Khairpur Mirs 66111, Pakistan; (A.G.S.); (A.T.); (T.P.)
| | - Baradi Waryani
- Department of Fresh Water Biology and Fisheries, University of Sindh, Jamshoro 76080, Pakistan;
| | | | - Tajnees Pirzada
- Institute of Chemistry, Shah Abdul Latif University Khairpur Mirs, Khairpur Mirs 66111, Pakistan; (A.G.S.); (A.T.); (T.P.)
| | - Ayman Nafady
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Elmuez A. Dawi
- Nonlinear Dynamics Research Centre (NDRC), Ajman University, Ajman P.O. Box 346, United Arab Emirates; (E.A.D.); (A.A.K.H.I.)
| | - Lama M. A. Saleem
- Biomolecular Science, Earth and Life Science, Amsterdam University, 1081 HV Amsterdam, The Netherlands;
| | - Mohsen Padervand
- Department of Chemistry, Faculty of Science, University of Maragheh, Maragheh P.O. Box. 55181-83111, Iran;
| | - Abd Al Karim Haj Ismail
- Nonlinear Dynamics Research Centre (NDRC), Ajman University, Ajman P.O. Box 346, United Arab Emirates; (E.A.D.); (A.A.K.H.I.)
| | - Kangle Lv
- College of Resource and Environment, South-Central Minzu University, Wuhan 430074, China;
| | - Brigitte Vigolo
- Institut Jean Lamour, Université de Lorraine, CNRS, IJL, F-54000 Nancy, France;
| | | |
Collapse
|
17
|
Zhang JZ, Shi NR, Wu JS, Wang X, Illes P, Tang Y. UDP-glucose sensing P2Y 14R: A novel target for inflammation. Neuropharmacology 2023; 238:109655. [PMID: 37423482 DOI: 10.1016/j.neuropharm.2023.109655] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/11/2023]
Abstract
Uridine 5'-diphosphoglucose (UDP-G) as a preferential agonist, but also other UDP-sugars, such as UDP galactose, function as extracellular signaling molecules under conditions of cell injury and apoptosis. Consequently, UDP-G is regarded to function as a damage-associated molecular pattern (DAMP), regulating immune responses. UDP-G promotes neutrophil recruitment, leading to the release of pro-inflammatory chemokines. As a potent endogenous agonist with the highest affinity for the P2Y14 receptor (R), it accomplishes an exclusive relationship between P2Y14Rs in regulating inflammation via cyclic adenosine monophosphate (cAMP), nod-like receptor protein 3 (NLRP3) inflammasome, mitogen-activated protein kinases (MAPKs), and signal transducer and activator of transcription 1 (STAT1) pathways. In this review, we initially present a brief introduction into the expression and function of P2Y14Rs in combination with UDP-G. Subsequently, we summarize emerging roles of UDP-G/P2Y14R signaling pathways that modulate inflammatory responses in diverse systems, and discuss the underlying mechanisms of P2Y14R activation in inflammation-related diseases. Moreover, we also refer to the applications as well as effects of novel agonists/antagonists of P2Y14Rs in inflammatory conditions. In conclusion, due to the role of the P2Y14R in the immune system and inflammatory pathways, it may represent a novel target for anti-inflammatory therapy.
Collapse
Affiliation(s)
- Ji-Zhou Zhang
- International Joint Research Centre on Purinergic Signalling, School of Acupuncture and Tuina/Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Nan-Rui Shi
- International Joint Research Centre on Purinergic Signalling, School of Acupuncture and Tuina/Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Jia-Si Wu
- International Joint Research Centre on Purinergic Signalling, School of Acupuncture and Tuina/Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Xin Wang
- International Joint Research Centre on Purinergic Signalling, School of Acupuncture and Tuina/Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Peter Illes
- International Joint Research Centre on Purinergic Signalling, School of Acupuncture and Tuina/Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China; Rudolf Boehm Institute for Pharmacology and Toxicology, University of Leipzig, 04107, Leipzig, Germany.
| | - Yong Tang
- International Joint Research Centre on Purinergic Signalling, School of Acupuncture and Tuina/Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China; Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, 610075, China.
| |
Collapse
|
18
|
Chandrakumar HP, Puskoor AV, Chillumuntala S, Sharif S, Mora DC, Gupta T, Tadayoni A, Zonnoor SL, Celiker P, Yakoubovitch S, Tsai A, Chiu C, Kazi A, Freilich M, Saleh A, Moon SJ, Cruickshank K, Li S, Katz J, Eichler J, Salciccioli L, McFarlane IM. Assessment of Cardiovascular Disease Among Predominantly Black Gout Patients. J Clin Rheumatol 2023; 29:202-206. [PMID: 36881837 DOI: 10.1097/rhu.0000000000001948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
INTRODUCTION Although the association between gout and cardiovascular disease (CVD) has been extensively studied, scarce data are available for the Black population. We aimed to assess the association between gout and CVD in a predominantly Black urban population with gout. METHODS A cross-sectional analysis was performed between a gout cohort and an age-/sex-matched control group. Clinical parameters and 2D echocardiograms were reviewed for the patients with gout and heart failure (HF). The primary outcome studied includes the prevalence and strength of association between gout and CVD. Secondary outcomes studied includes strength of association of gout and HF categorized by ejection fraction, mortality, and HF readmissions. RESULTS Four hundred seventy-one patients with gout had a mean age of 63.7 ± 0.5 years; 89% were Black, 63% were men, and mean body mass index was 31.3 ± 0.4 kg/m 2 . Hypertension, diabetes mellitus, and dyslipidemia were present in 89%, 46%, and 52%, respectively. Compared with controls, patients with gout had significantly higher rates of angina, arrhythmia, coronary artery disease/stents, myocardial infarction, coronary artery bypass graft surgery, cerebrovascular accident, and peripheral vascular disease. The adjusted odds ratio for CVD was 2.9 (95% confidence interval, 1.9-4.5; p < 0.001). Gout patients had a higher prevalence of HF with 45% (n = 212) compared with controls with 9.4% (n = 44). Adjusted odds ratio for HF risk was 7.1 (95% confidence interval, 4.7-10.6; p < 0.01). CONCLUSIONS Gout in a predominantly Black population confers 3 times the CVD risk and 7 times HF-specific risk compared with age- and sex-matched cohort. Further research is needed to confirm our findings and to develop interventions to reduce morbidity associated with gout.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Louis Salciccioli
- Department of Cardiology, SUNY Downstate Health Sciences University, Brooklyn, NY
| | | |
Collapse
|
19
|
Charoenwutthikun S, Chanjitwiriya K, Roytrakul S, Kunthalert D. A wild rice-derived peptide R14 ameliorates monosodium urate crystals-induced IL-1β secretion through inhibition of NF-κB signaling and NLRP3 inflammasome activation. PeerJ 2023; 11:e15295. [PMID: 37197585 PMCID: PMC10184658 DOI: 10.7717/peerj.15295] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 04/04/2023] [Indexed: 05/19/2023] Open
Abstract
Gout is an inflammatory arthritis initiated by the deposition of monosodium urate crystals (MSU) around the joints and surrounding tissues. MSU crystals activate the nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasome to the release of interleukin-1β (IL-1β). Gout can have a substantial impact on patient's quality of life, and currently available medicines are unable to meet all the clinical needs. This study explored anti-gout potentials of the Rice14 (R14) peptide, a peptide derived from leaves of wild rice Oryza minuta. The effects of R14 peptide on IL-1β secretion in THP-1 macrophages with MSU crystals-induced inflammation were examined. Our results clearly showed that the R14 peptide significantly inhibited the secretion of IL-1β in MSU crystals-induced macrophages, and the effects were dose-related. For safety testing, the R14 peptide did not show both cytotoxicity and hemolytic activity. In addition, the R14 peptide strongly suppressed the phospho-IκB-α and nuclear factor kappa-B (NF-κB) p65 proteins in NF-κB signaling pathway, reduced the NLRP3 expression and inhibited the MSU crystals-mediated cleavage of caspase-1 as well as mature IL-1β. The R14 peptide also reduced MSU-triggered intracellular ROS levels in macrophages. Taken together, these results indicated that R14 peptide inhibited MSU crystals-induced IL-1β production through NF-κB and NLRP3 inflammasome activation. Our findings demonstrated that R14 peptide, the newly recognized peptide from wild rice, possessed potent regulatory activity against IL-1β production in MSU crystals-induced inflammation, and we therefore propose that the R14 peptide is a promising molecule with potential clinical application in the treatment of MSU crystals-induced inflammation.
Collapse
Affiliation(s)
- Supattra Charoenwutthikun
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Kasem Chanjitwiriya
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Sittiruk Roytrakul
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Thailand Science Park, Pathumthani, Thailand
| | - Duangkamol Kunthalert
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
- Centre of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| |
Collapse
|
20
|
Xu H, Zhang B, Chen Y, Zeng F, Wang W, Chen Z, Cao L, Shi J, Chen J, Zhu X, Xue Y, He R, Ji M, Hua Y. Type II collagen facilitates gouty arthritis by regulating MSU crystallisation and inflammatory cell recruitment. Ann Rheum Dis 2023; 82:416-427. [PMID: 36109143 DOI: 10.1136/ard-2022-222764] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/03/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Increasing evidence suggests that impaired cartilage is a substantial risk factor for the progression from hyperuricaemia to gout. Since the relationship between cartilage matrix protein and gout flares remains unclear, we investigated its role in monosodium urate (MSU) crystallisation and following inflammation. METHODS Briefly, we screened for cartilage matrix in synovial fluid from gouty arthritis patients with cartilage injuries. After identifying a correlation between crystals and matrix molecules, we conducted image analysis and classification of crystal phenotypes according to their morphology. We then evaluated the differences between the cartilage matrix protein-MSU complex and the pure MSU crystal in their interaction with immune cells and identified the related signalling pathway. RESULTS Type II collagen (CII) was found to be enriched around MSU crystals in synovial fluid after cartilage injury. Imaging analysis revealed that CII regulated the morphology of single crystals and the alignment of crystal bows in the co-crystalline system, leading to greater phagocytosis and oxidative stress in macrophages. Furthermore, CII upregulated MSU-induced chemokine and proinflammatory cytokine expression in macrophages, thereby promoting the recruitment of leucocytes. Mechanistically, CII enhanced MSU-mediated inflammation by activating the integrin β1(ITGB1)-dependent TLR2/4-NF-κB signal pathway. CONCLUSION Our study demonstrates that the release of CII and protein-crystal adsorption modifies the crystal profile and promotes the early immune response in MSU-mediated inflammation. These findings open up a new path for understanding the relationship between cartilage injuries and the early immune response in gout flares.
Collapse
Affiliation(s)
- HanLin Xu
- Department of Sports Medicine, Huashan Hospital,Fudan University, Shanghai, China
| | - Bohan Zhang
- State Key Laboratory of Surface Physics and Department of Physics, Human Phenome Institute, Academy for Engineering and Technology, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Yiwu Research Institute, Fudan University, Shanghai, China
| | - Yaxin Chen
- State Key Laboratory of Surface Physics and Department of Physics, Human Phenome Institute, Academy for Engineering and Technology, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Yiwu Research Institute, Fudan University, Shanghai, China
| | - Fengzhen Zeng
- School of Communication and Information Engineering, Shanghai University, Shanghai, China
| | - Wenjuan Wang
- Department of Sports Medicine, Huashan Hospital,Fudan University, Shanghai, China
| | - Ziyi Chen
- Department of Sports Medicine, Huashan Hospital,Fudan University, Shanghai, China
| | - Ling Cao
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jun Shi
- School of Communication and Information Engineering, Shanghai University, Shanghai, China
| | - Jun Chen
- Department of Sports Medicine, Huashan Hospital,Fudan University, Shanghai, China
| | - Xiaoxia Zhu
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu Xue
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Rui He
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China .,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - MinBiao Ji
- State Key Laboratory of Surface Physics and Department of Physics, Human Phenome Institute, Academy for Engineering and Technology, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Yiwu Research Institute, Fudan University, Shanghai, China
| | - YingHui Hua
- Department of Sports Medicine, Huashan Hospital,Fudan University, Shanghai, China
| |
Collapse
|
21
|
Liu Y, Zhu X, Ji S, Huang Z, Zang Y, Ding Y, Zhang J, Ding Z. Transdermal delivery of colchicine using dissolvable microneedle arrays for the treatment of acute gout in a rat model. Drug Deliv 2022; 29:2984-2994. [PMID: 36101018 PMCID: PMC9487926 DOI: 10.1080/10717544.2022.2122632] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Colchicine (Col) is used to prevent and treat acute gout flare; however, its therapeutic use is strictly limited owing to severe gastrointestinal side effects after oral administration. Therefore, we developed a dissolvable Col-loaded microneedle (MN) with hyaluronic acid to deliver Col via the transdermal route. We studied the preparation, mechanical properties, skin insertion, skin irritation, drug content, and transdermal release of the Col-loaded MN. The pharmacokinetics of Col after Col-loaded MN application were compared with those of Col solution gavage over 24 h. Knee joint edema evaluation and the hindfoot mechanical threshold test were conducted to determine the pharmacodynamic profile. The dissolvable Col-loaded MN possessed sufficient mechanical strength to penetrate the skin and release the loaded drug. No skin irritation was observed for 3 days after application. We found that 3.36-fold more Col contained in MNs was delivered through the skin compared with that in gel in vitro, and moderate relative bioavailability in vivo. The Col-loaded MN significantly relieved swollen knee joints and mechanical hypernociception in an acute gout model in rats. The dissolvable Col-loaded MN array reduced inflammation and pain via topical administration when acute gout occurred. Reducing the gastrointestinal side effects of Col-loaded MNs is expected to optimize the therapeutic effects of Col and improve patient compliance.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Xiaoruo Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Shiliang Ji
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
- Department of Pharmacy, Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Zhen Huang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yuhui Zang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Ying Ding
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Junfeng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Zhi Ding
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
- Engineering Research Center of Protein and Peptide Medicine, Ministry of Education, Nanjing, China
- Changzhou High-Tech Research Institute of Nanjing University, Changzhou, China
| |
Collapse
|
22
|
Wikan N, Potikanond S, Hankittichai P, Thaklaewphan P, Monkaew S, Smith DR, Nimlamool W. Alpinetin Suppresses Zika Virus-Induced Interleukin-1β Production and Secretion in Human Macrophages. Pharmaceutics 2022; 14:pharmaceutics14122800. [PMID: 36559293 PMCID: PMC9782830 DOI: 10.3390/pharmaceutics14122800] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/08/2022] [Accepted: 12/11/2022] [Indexed: 12/16/2022] Open
Abstract
Zika virus (ZIKV) infection has been recognized to cause adverse sequelae in the developing fetus. Specially, this virus activates the excessive release of IL-1β causing inflammation and altered physiological functions in multiple organs. Although many attempts have been invested to develop vaccine, antiviral, and antibody therapies, development of agents focusing on limiting ZIKV-induced IL-1β release have not gained much attention. We aimed to study the effects of alpinetin (AP) on IL-1β production in human macrophage upon exposure to ZIKV. Our study demonstrated that ZIKV stimulated IL-1β release in the culture supernatant of ZIKV-infected cells, and AP could effectively reduce the level of this cytokine. AP exhibited no virucidal activities against ZIKV nor caused alteration in viral production. Instead, AP greatly inhibited intracellular IL-1β synthesis. Surprisingly, this compound did not inhibit ZIKV-induced activation of NF-κB and its nuclear translocation. However, AP could significantly inhibit ZIKV-induced p38 MAPK activation without affecting the phosphorylation status of ERK1/2 and JNK. These observations suggest the possibility that AP may reduce IL-1β production, in part, through suppressing p38 MAPK signaling. Our current study sheds light on the possibility of using AP as an alternative agent for treating complications caused by ZIKV infection-induced IL-1β secretion.
Collapse
Affiliation(s)
- Nitwara Wikan
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand
| | - Saranyapin Potikanond
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Research Center for Development of Local Lanna Rice and Rice Products, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Phateep Hankittichai
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Phatarawat Thaklaewphan
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sathit Monkaew
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Duncan R. Smith
- Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand
- Correspondence: (D.R.S.); (W.N.); Tel.: +66-53-934597 (W.N.)
| | - Wutigri Nimlamool
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Research Center for Development of Local Lanna Rice and Rice Products, Chiang Mai University, Chiang Mai 50200, Thailand
- Correspondence: (D.R.S.); (W.N.); Tel.: +66-53-934597 (W.N.)
| |
Collapse
|
23
|
Zhang W, Chen L, Lou M. Association of Elevated Serum Uric Acid with Nerve Conduction Function and Peripheral Neuropathy Stratified by Gender and Age in Type 2 Diabetes Patients. Brain Sci 2022; 12:brainsci12121704. [PMID: 36552164 PMCID: PMC9775627 DOI: 10.3390/brainsci12121704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Background: The relationship between serum uric acid (SUA) level and diabetic peripheral neuropathy (DPN) remains controversial. We aimed to investigate the association between SUA level and DPN and evaluate the effects of SUA level on nerve conduction function via electromyography in patients with type 2 diabetes (T2DM), stratified by gender and age. Methods: This cross-sectional study included 647 inpatients with T2DM from the First Affiliated Hospital of Wenzhou Medical University between February 2017 and October 2020. The diagnosis of DPN was confirmed according to the Toronto Expert Consensus. Clinical data, SUA level, and nerve conduction parameters were obtained from electronic medical records. Results: A total of 647 patients with T2DM were included, and 471 patients were diagnosed with DPN. The level of SUA was higher in the DPN group than in the Non-DPN group (330.58 ± 99.67 vs. 309.16 ± 87.04, p < 0.05). After adjustment, a higher SUA level was associated with the presence of DPN [odds ratio (OR) 1.003, 95% confidence interval (CI), 1.001−1.005; p = 0.017]. The area under the curve for the prediction of DPN was 0.558 (95% CI, 0.509−0.608; p = 0.022), and the optimized cut-off of SUA level was 297.5 µmol/L. The SUA > 297.5 µmol/L level was independently associated with DPN in the male subgroup (OR 2.507, 95% CI, 1.405−4.473; p = 0.002) rather than in the female subgroup. Besides, SUA > 297.5 µmol/L was independently associated with DPN in the younger subgroup (age < 65 years) (OR 2.070, 95% CI, 1.278−3.352; p = 0.003) rather than in the older subgroup. In multiple linear regression analysis, SUA was significantly correlated with certain nerve conduction study parameters in the all patients group, and was also observed in the male and younger subgroups. Conclusions: Elevated SUA was independently associated with poorer nerve conduction functions, and hyperuricemia was also significantly associated with a higher risk of developing DPN in T2DM patients, especially in male and younger patients.
Collapse
Affiliation(s)
- Wanli Zhang
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310009, China
| | - Lingli Chen
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Min Lou
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310009, China
- Correspondence: ; Tel.: +86-571-87784810; Fax: +86-571-87784850
| |
Collapse
|
24
|
Cao Y, Zhong Q, Tang F, Yao X, Liu Z, Zhang X. Anethole ameliorates inflammation induced by monosodium urate in an acute gouty arthritis model via inhibiting TLRs/MyD88 pathway. Allergol Immunopathol (Madr) 2022; 50:107-114. [PMID: 36335453 DOI: 10.15586/aei.v50i6.682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 06/09/2022] [Indexed: 06/16/2023]
Abstract
OBJECTIVE To assess the effects of anethole on monosodium urate (MSU)-induced inflammatory response, investigate its role in acute gouty arthritis (AGA), and verify its molecular mechanism. METHODS Hematoxylin and eosin staining assay and time-dependent detection of degree of ankle swelling were performed to assess the effects of anethole on joint injury in MSU-induced AGA mice. Enzyme-linked-immunosorbent serologic assay was performed to demonstrate the production levels of inflammatory factors (interleukin 1β [IL-1β], interleukin 6 [IL-6], interleukin 8 [IL-8], tumor necrosis factor α [TNF-α], and monocyte chemo-attractant protein-1 [MCP-1]) in MSU-induced AGA mice. Western blot assays were used to confirm the effects of anethole on oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome activity and the activation of toll-like receptors (TLRs)-myeloid differentiation factor 88 (MyD88) pathway in MSU-induced AGA mice. RESULTS We observed that a significant joint injury occurred in MSU-induced AGA mice. Anethole could alleviate the pathological injury of the synovium in MSU-induced AGA mice and suppressed ankle swelling. In addition, we observed that anethole could inhibit MSU-induced inflammatory response and inflammasome activation in MSU-induced AGA mice. Moreover, we discovered that anethole enabled to inhibit the activation of TLRs/MyD88 pathway in MSU-induced AGA mice. Our findings further confirmed that anethole contributed to the inhibitory effects on progression in MSU-induced AGA mice. CONCLUSION It confirmed that anethole ameliorated the MSU-induced inflammatory response in AGA mice in vivo via inhibiting TLRs-MyD88 pathway.
Collapse
Affiliation(s)
- Yuepeng Cao
- Department of Rheumatology and Immunology, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Qin Zhong
- Department of Rheumatology and Immunology, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Fang Tang
- Department of Rheumatology and Immunology, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Xueming Yao
- Department of Rheumatology and Immunology, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Zhengqi Liu
- Department of Rheumatology and Immunology, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China;
| | - Xiaodong Zhang
- Second Clinical School of Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| |
Collapse
|
25
|
Tian J, Zhou D, Xiang L, Xie B, Wang B, Li Y, Liu X. Calycosin represses AIM2 inflammasome-mediated inflammation and pyroptosis to attenuate monosodium urate-induced gouty arthritis through NF-κB and p62-Keap1 pathways. Drug Dev Res 2022; 83:1654-1672. [PMID: 36069386 DOI: 10.1002/ddr.21985] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 07/05/2022] [Accepted: 08/10/2022] [Indexed: 11/06/2022]
Abstract
Gouty arthritis is an inflammatory disease induced by monosodium urate (MSU), and is closely related to the activation of inflammasomes. Calycosin plays an anti-inflammatory role in arthritis. This study explored the mechanism of Calycosin in MSU-induced gouty arthritis. MSU-induced gouty arthritis mouse models with or without treatment of Calycosin were established, and physiological and pathological indicators were determined. Similarly, peripheral blood mononuclear cells (PBMCs) and THP-1 macrophages were used in vitro. Lactate dehydrogenase (LDH) was tested. The degree of centrifugal infiltration was detected by immunofluorescence. ELISA and quantitative reverse-transcription polymerase chain reaction were conducted to determine the levels of inflammatory factors. Immunohistochemistry, immunofluorescence, and flow cytometry were utilized to detect the content of caspase-1. Protein expressions of NF-κB-, p62-Keap1 pathway-, and pyroptosis-related factors were examined by western blot. In MSU-induced mouse models, calycosin increased mechanical hyperalgesia but decreased the swelling index of the mouse knee joint in a time-dependent manner. MSU treatment increased inflammatory cells and LysM-eGFP+ neutrophils recruitment in vivo, and promoted the LDH content in vitro, and meanwhile, calycosin reversed the aforementioned effects of MSU. In addition, calycosin repressed the release of inflammatory factors, promoted p62 level and diminished the levels of AIM2, caspase-1, ASC, IL-1β, Keap1, Cleaved GSDMD, and Cleaved caspase-1 and phosphorylation of p65 and IκBα in MSU-induced mouse or cell models. Furthermore, AIM2 silencing also inhibited MSU-induced inflammation and pyroptosis. Collectively, calycosin may inhibit AIM2 inflammasomes-mediated inflammation and pyroptosis through NF-κB and p62-Keap1 pathways, ultimately playing a protective role in gouty arthritis.
Collapse
Affiliation(s)
- Jing Tian
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, China
| | - Dapeng Zhou
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, China
| | - Liangbi Xiang
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, China
| | - Bing Xie
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, China
| | - Baichuan Wang
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, China
| | - Yang Li
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, China
| | - Xinwei Liu
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
26
|
Fernández-Torres J, Martínez-Nava GA, Martínez-Flores K, Sánchez-Sánchez R, Jara LJ, Zamudio-Cuevas Y. The interplay between HLA-B and NLRP3 polymorphisms may be associated with the genetic susceptibility of gout. Mol Biol Rep 2022; 49:10205-10215. [PMID: 36057006 DOI: 10.1007/s11033-022-07895-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/22/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND HLA and NLRP3 play an important role in the development of various autoimmune and autoinflammatory diseases. Gout is an autoinflammatory disease associated with multiple genetic and environmental factors. The objective of the present study was to evaluate the interaction and association between genetic polymorphisms of HLA-B and the NLRP3 gene in Mexican patients with gout. METHODS AND RESULTS Eighty-one patients with gout were included and compared with 95 healthy subjects. The polymorphisms rs4349859, rs116488202, rs2734583 and rs3099844 (within the HLA-B region) and rs3806268 and rs10754558 of the NLRP3 gene were genotyped using TaqMan probes in a Rotor-Gene device. The interactions were determined using the multifactorial dimensionality reduction (MDR) method, while the associations were determined through logistic regression models. The MDR analysis revealed significant interactions between the rs116488202 and rs10754558 polymorphisms with an entropy value of 4.31% (p < 0.0001). Significant risk associations were observed with rs4349859 and rs116488202 polymorphisms (p < 0.01); however, no significant associations were observed with the polymorphisms of the NLRP3 gene. CONCLUSIONS The results suggest that HLA-B polymorphisms and their interaction with NLRP3 may contribute to the genetic susceptibility of gout.
Collapse
Affiliation(s)
- Javier Fernández-Torres
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Mexico City, Mexico.,Biology Department, Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | | | - Karina Martínez-Flores
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Mexico City, Mexico
| | - Roberto Sánchez-Sánchez
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Mexico City, Mexico
| | - Luis J Jara
- Rheumatology Division, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Mexico City, Mexico
| | - Yessica Zamudio-Cuevas
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Mexico City, Mexico.
| |
Collapse
|
27
|
Targeting of Nrf2/PPARγ/NLRP3 Signaling Pathway by Stevia rebudiana Bertoni Extract Provides a Novel Insight into Its Protective Effect against Acute Gouty Arthritis-Induced Synovial Inflammation, Oxidative Stress and Apoptosis in a Rat Model. Processes (Basel) 2022. [DOI: 10.3390/pr10091751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Our research work examined the potential protection of Stevia rebaudiana extract against monosodium urate crystals (MSU)-induced acute gouty arthritis in a rat model and its possible underlying mechanism. Forty rats were allocated into four groups (n = 10); a control group; an MSU group, whose rats received 0.1 of MSU single intra-articular injection in the ankle joint on the fifth day of the experiment; an MSU + Stevia group, which received 250 mg/kg/day of Stevia extract orally for seven days and MSU crystals on the fifth day; and an MSU + colchicine group, which was administered colchicine at 0.28 mg/kg daily for seven days and MSU crystals on the fifth day. Pretreatment with Stevia extract mitigated MSU-induced inflammation as evidenced by a decrease of the ankle edema and inflammatory cell infiltration and a significant downregulation of the protein level of NFκB, TNFα, IL-1β, IL6, and IL18 as well as NLRP3 gene expression. Additionally, there was a markedly increased PPARγ gene expression (p < 0.001) compared with the MSU group (p < 0.001) and alleviated oxidative stress via significant upregulating of Nrf2/HO-1. Moreover, the pretreatment attenuated apoptosis by significantly decreasing cytochrome c, Bax, Caspase-3, and by increasing Bcl-2 protein. In conclusion, Stevia extract exhibited strong anti-inflammatory, antioxidant, and antiapoptotic effects against MSU-induced gouty arthritis similar to the standard anti-inflammatory colchicine drugs.
Collapse
|
28
|
Narendra Kumar AV, Muthu Prabhu S, Shin WS, Yadav KK, Ahn Y, Abdellattif MH, Jeon BH. Prospects of non-noble metal single atoms embedded in two-dimensional (2D) carbon and non-carbon-based structures in electrocatalytic applications. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214613] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
29
|
Feng W, Zhong XQ, Zheng XX, Liu QP, Liu MY, Liu XB, Lin CS, Xu Q. Study on the effect and mechanism of quercetin in treating gout arthritis. Int Immunopharmacol 2022; 111:109112. [PMID: 35932610 DOI: 10.1016/j.intimp.2022.109112] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 07/19/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022]
Abstract
Quercetin is widely found in natural plants, especially Chinese herbal plants. It has been used to treat arthritis in China for thousands of years. However, the effects and mechanisms of quercetin in the treatment of gout arthritis (GA) remain unclear. We aimed to verify the treatment of GA with quercetin and investigate the underlying mechanism. A combination of network pharmacology and experiments was used to reveal the mechanism of quercetin in the treatment of GA. Potential targets of quercetin and gout were identified. Then, the protein-protein interaction network for the common targets between quercetin and gout was constructed and the core targets were identified. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses for the common targets were performed to elucidate the pharmacological functions and mechanisms associated with quercetin treatment in GA. Finally, a monosodium urate-induced GA rat model was used to validate the predicted mechanisms in network pharmacology. Seventy-two common targets were identified. KEGG analysis revealed that treatment of GA with quercetin predominantly involved the interleukin (IL)-17, tumor necrosis factor (TNF), mitogen-activated protein kinase, and phosphoinositide 3-kinase-Akt signaling pathways. In an experimental validation, quercetin attenuated ankle joint inflammation-induced bone destruction and histological lesions. It also diminished the expression of IL-6, IL-17A, and IL-17F in the IL-17 pathway, and regulated the release of RAR-related orphan receptor gamma t,IL-17E, IL-1β, IL-6, TNF-α, Foxp3, and transforming growth factor-beta 1. The collective findings implicate quercetin as a valuable alternative drug for the treatment of GA.
Collapse
Affiliation(s)
- Wei Feng
- The First Clinical Medicine School, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xiao-Qin Zhong
- The First Clinical Medicine School, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xue-Xia Zheng
- The First Clinical Medicine School, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Qing-Ping Liu
- The First Clinical Medicine School, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Min-Ying Liu
- The First Clinical Medicine School, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xiao-Bao Liu
- The First Clinical Medicine School, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Chang-Song Lin
- The First Clinical Medicine School, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Qiang Xu
- The First Clinical Medicine School, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| |
Collapse
|
30
|
Santos PD, Vieira TN, Gontijo Couto AC, Mesquita Luiz JP, Lopes Saraiva AL, Borges Linhares CR, Barbosa MF, Justino AB, Franco RR, da Silva Brum E, Oliveira SM, Dechichi P, Pivatto M, de Melo Rodrigues Ávila V, Espíndola FS, Silva CR. Stephalagine, an aporphinic alkaloid with therapeutic effects in acute gout arthritis in mice. JOURNAL OF ETHNOPHARMACOLOGY 2022; 293:115291. [PMID: 35427727 DOI: 10.1016/j.jep.2022.115291] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/23/2022] [Accepted: 04/08/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Gout is an inflammatory disease characterized by the accumulation of monosodium urate crystals (MSU) in the joints, leading to severe pain and inflammation. Stephalagine is a Brazilian Savanna aporphine alkaloid isolated from Annona crassiflora Mart. Fruit peel, that has been popularly used to treat rheumatism and have been described with antinociceptive properties. However, no studies evaluated the possible therapeutic properties of stephalagine in arthritic pain. AIM OF THE STUDY To evaluate the possible antinociceptive and anti-inflammatory effects of stephalagine in an acute gout attack in mice. MATERIALS AND METHODS Adult male wild type C57BL/6/J/UFU mice (20-25 g) were used (process number 018/17). The treated group received stephalagine (1 mg/kg, by gavage) and the vehicle group received saline (10 mL/kg, by gavage), both 1 h before the MSU crystals (100 μg/ankle joint) administration. All groups were analyzed for mechanical allodynia, thermal hyperalgesia, overt pain-like behaviors, and edema development at 2, 4, 6 and 24 h after injections. Synovial fluid and the ankle articulation from the injected joint were collected 4 h after administrations for myeloperoxidase enzyme activity, IL-1β measurement, and histological analysis. RESULTS Stephalagine had a significant antinociceptive effect on mechanical allodynia, when compared to vehicle group at 2-24 h after intra-articular injection of MSU and 2 h for spontaneous and cold thermal sensitivity. Stephalagine was also able to significantly reduce the articular edema (45 ± 1%), the activity of the myeloperoxidase enzyme (37 ± 6%), and IL-1β levels (43 ± 3%). The histological analysis confirms that stephalagine dramatically reduced the number of infiltrating inflammatory cells (75 ± 6%) in MSU injected animals. Also, stephalagine treatment did not alter the uric acid levels, xanthine oxidase activity, AST and ALT activities, urea and creatinine levels, neither cause any macroscopic changes in the mice's weight, deformations, changes in the coat, or feces. CONCLUSION Stephalagine may be an alternative for the management of gout, once it was able to induce antinociceptive and anti-inflammatory effects without causing adverse effects on the evaluated parameters.
Collapse
Affiliation(s)
- Priscilla Dias Santos
- Graduate Program in Genetics and Biochemistry, Institute of Biotechnology, Federal University of Uberlândia, 38408-100, Uberlândia, (MG), Brazil.
| | - Thiago Neves Vieira
- Graduate Program in Genetics and Biochemistry, Institute of Biotechnology, Federal University of Uberlândia, 38408-100, Uberlândia, (MG), Brazil
| | - Ana Claudia Gontijo Couto
- Graduate Program in Genetics and Biochemistry, Institute of Biotechnology, Federal University of Uberlândia, 38408-100, Uberlândia, (MG), Brazil
| | - João Paulo Mesquita Luiz
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 14049-900, Ribeirão Preto, (SP), Brazil
| | - André Luis Lopes Saraiva
- Graduate Program in Genetics and Biochemistry, Institute of Biotechnology, Federal University of Uberlândia, 38408-100, Uberlândia, (MG), Brazil
| | | | - Marília Fontes Barbosa
- Nucleus of Research on Bioactive Compounds (NPCBio), Institute of Chemistry, Federal University of Uberlândia, 38408-100, Uberlândia, (MG), Brazil
| | - Allisson Benatti Justino
- Graduate Program in Genetics and Biochemistry, Institute of Biotechnology, Federal University of Uberlândia, 38408-100, Uberlândia, (MG), Brazil
| | - Rodrigo Rodrigues Franco
- Graduate Program in Genetics and Biochemistry, Institute of Biotechnology, Federal University of Uberlândia, 38408-100, Uberlândia, (MG), Brazil
| | - Evelyne da Silva Brum
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria (UFSM), 97105-900, Santa Maria, (RS), Brazil
| | - Sara Marchesan Oliveira
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria (UFSM), 97105-900, Santa Maria, (RS), Brazil
| | - Paula Dechichi
- Department of Cellular Biology, Histology and Embryology, Institute of Biomedical Sciences, Federal University of Uberlândia, 38400-902, Uberlândia, (MG), Brazil
| | - Marcos Pivatto
- Nucleus of Research on Bioactive Compounds (NPCBio), Institute of Chemistry, Federal University of Uberlândia, 38408-100, Uberlândia, (MG), Brazil
| | - Veridiana de Melo Rodrigues Ávila
- Graduate Program in Genetics and Biochemistry, Institute of Biotechnology, Federal University of Uberlândia, 38408-100, Uberlândia, (MG), Brazil
| | - Foued Salmen Espíndola
- Graduate Program in Genetics and Biochemistry, Institute of Biotechnology, Federal University of Uberlândia, 38408-100, Uberlândia, (MG), Brazil
| | - Cássia Regina Silva
- Graduate Program in Genetics and Biochemistry, Institute of Biotechnology, Federal University of Uberlândia, 38408-100, Uberlândia, (MG), Brazil.
| |
Collapse
|
31
|
Dempsey B, Cruz LC, Mineiro MF, da Silva RP, Meotti FC. Uric Acid Reacts with Peroxidasin, Decreases Collagen IV Crosslink, Impairs Human Endothelial Cell Migration and Adhesion. Antioxidants (Basel) 2022; 11:antiox11061117. [PMID: 35740014 PMCID: PMC9220231 DOI: 10.3390/antiox11061117] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 02/04/2023] Open
Abstract
Uric acid is considered the main substrate for peroxidases in plasma. The oxidation of uric acid by human peroxidases generates urate free radical and urate hydroperoxide, which might affect endothelial function and explain, at least in part, the harmful effects of uric acid on the vascular system. Peroxidasin (PXDN), the most recent heme-peroxidase described in humans, catalyzes the formation of hypobromous acid, which mediates collagen IV crosslinks in the extracellular matrix. This enzyme has gained increasing scientific interest since it is associated with cardiovascular disease, cancer, and renal fibrosis. The main objective here was to investigate whether uric acid would react with PXDN and compromise the function of the enzyme in human endothelial cells. Urate decreased Amplex Red oxidation and brominating activity in the extracellular matrix (ECM) from HEK293/PXDN overexpressing cells and in the secretome of HUVECs. Parallelly, urate was oxidized to 5-hydroxyisourate. It also decreased collagen IV crosslink in isolated ECM from PFHR9 cells. Urate, the PXDN inhibitor phloroglucinol, and the PXDN knockdown impaired migration and adhesion of HUVECs. These results demonstrated that uric acid can affect extracellular matrix formation by competing for PXDN. The oxidation of uric acid by PXDN is likely a relevant mechanism in the endothelial dysfunction related to this metabolite.
Collapse
|
32
|
Riaz M, Al Kury LT, Atzaz N, Alattar A, Alshaman R, Shah FA, Li S. Carvacrol Alleviates Hyperuricemia-Induced Oxidative Stress and Inflammation by Modulating the NLRP3/NF-κB Pathwayt. Drug Des Devel Ther 2022; 16:1159-1170. [PMID: 35496367 PMCID: PMC9041362 DOI: 10.2147/dddt.s343978] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 03/22/2022] [Indexed: 12/20/2022] Open
Abstract
Purpose Gouty arthritis is generally induced by the accumulation of monosodium urate (MSU) crystals in the joints due to elevated serum uric acid levels, potentially leading to serious pathological disorders such as nephrolithiasis, renal failure, and acute gouty arthritis. In this study, we aimed to validate the anti-gout effects of carvacrol, a phenolic monoterpene. Materials and Methods Male Sprague–Dawley rats were divided into normal saline, disease group by injecting potassium mono-oxonate (PO) at a dose of 250 mg/kg, and three treatment groups, either with carvacrol 20 mg/kg or 50 mg/kg and 10 mg/kg allopurinol. The blood and tissue samples were subsequently collected and analyzed using different biochemical and histopathological techniques. Results Our results revealed a significant increase in the serum levels of oxidative stress-related markers, namely, uric acid and C-reactive protein (CRP), and NLRP3 inflammasome-dependent inflammatory mediators, including nuclear factor kappa B (NF-κB) and tumor necrosis factor-alpha (TNF-α). Carvacrol administration for seven consecutive days exhibited significant anti-hyperuricemic and anti-inflammatory effects in a dose-dependent manner. Notably, the 50 mg/kg carvacrol treatment was observed to produce results similar to the allopurinol treatment. Furthermore, the renal safety of carvacrol was confirmed by the renal function test. Conclusion Carvacrol potentially alleviates hyperuricemia-induced oxidative stress and inflammation by regulating the ROS/NRLP3/NF-κB pathway, thereby exerting protective effects against joint degeneration.
Collapse
Affiliation(s)
- Muhammad Riaz
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Lina Tariq Al Kury
- Department of Natural and Health Sciences Zayed University, Abu Dhabi, United Arab Emirates
| | - Noreen Atzaz
- Department of Pathology, Benazir Bhutto Hospital, Rawalpindi, Pakistan
| | - Abdullah Alattar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
| | - Reem Alshaman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
| | - Fawad Ali Shah
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen, Shenzhen, People's Republic of China
| |
Collapse
|
33
|
Anti-inflammatory and Antioxidant Effect of Poly-gallic Acid (PGAL) in an In Vitro Model of Synovitis Induced by Monosodium Urate Crystals. Inflammation 2022; 45:2066-2077. [PMID: 35505045 DOI: 10.1007/s10753-022-01676-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/24/2022] [Accepted: 04/21/2022] [Indexed: 11/05/2022]
Abstract
Gout is a chronic and degenerative disease that affects the joints and soft tissues because of the crystalline deposit of monosodium urate. The interaction between monosodium urate crystals (MSU) and synoviocytes generates oxidative and inflammatory states. These physiological characteristics have promoted the study of poly-gallic acid (PGAL), a poly-oxidized form of gallic acid reported to be effective in in vitro models of inflammation. The effect of PGAL in an in vitro model of oxidation and synovial inflammation induced by MSU was evaluated after 24 h of stimulation through the morphological changes, the determination of oxidative stress (OS), IL-1β, and the phagocytosis of the MSU. A 20% reduction in synovial viability and the generation of vesicles were observed when they were exposed to MSU. When PGAL was used at 100 and 200 µg/ml, cell death was reduced by 30% and 17%, respectively. PGAL both doses reduce the vesicles generated by MSU. OS generation in synoviocytes exposed to 100 µg/ml and 200 µg/ml PGAL decreased by 1.28 and 1.46 arbitrary fluorescence units (AFU), respectively, compared to the OS in synoviocytes exposed to MSU (1.9 AFU). PGAL at 200 µg/ml inhibited IL-1β by 100%, while PGAL at 100 µg/ml inhibited IL-1β by 66%. The intracellular MSU decreased in synoviocytes stimulated with 100 µg/ml PGAL. The PGAL has a cytoprotective effect against damage caused by MSU in synoviocytes and can counteract the oxidative and inflammatory response induced by the crystals probably because it exerts actions at the membrane level that prevent phagocytosis of the crystals.
Collapse
|
34
|
Xian G, Wang L, Wan X, Yan H, Cheng J, Chen Y, Lu J, Li Y, Li D, Dou J, Wang S. Two Multiresponsive Luminescent Zn-MOFs for the Detection of Different Chemicals in Simulated Urine and Antibiotics/Cations/Anions in Aqueous Media. Inorg Chem 2022; 61:7238-7250. [PMID: 35504023 DOI: 10.1021/acs.inorgchem.1c03502] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Two Zn-MOFs, namely, {[Zn(L)0.5(bpea)]·0.5H2O·0.5DMF}n [LCU-113 (for Liaocheng University)] and {[Zn(L)0.5(ibpt)]·H2O·DMF}n (LCU-114), were synthesized based on flexible tetracarboxylic acid 1,3-bis(3,5-dicarboxyphenoxy)benzene (H4L) and different N-ligands [bpea = 1,2-dipyridyl ethane; ibpt = 3-(4'-imidazolobenzene)-5-(pyridine-4'-yl)-1,2,4-triazole]. LCU-113 and LCU-114 possess twofold interpenetrating three-dimensional pillared layer structures, in which a two-dimensional layer formed by carboxylic acid and Zn2+ ions was pillared by bpea and ibpt, respectively. The two complexes show high water stability and high luminescence sensing performance toward organic solvents, ions, and antibiotics, as well as chemicals, in simulated urine. The investigation showed that (1) LCU-113 and LCU-114 could detect uric acid (UA, 2,6,8-trihydroxypurine, metabolite of purine) and p-aminophenol (PAP, biomarker of phenamine) in simulated urine by luminescence quenching, respectively, and (2) luminescence quenching of LCU-113 and LCU-114 occurred in aqueous solutions of nitrofurazone (NZF), Fe3+, and CrO42-/Cr2O72-. All the above detections have excellent anti-interference ability and recyclability. The luminescence mechanism analysis indicates that weak interactions between the framework structures and the target analytes as well as the energy competition (inner filter effect) play an important role in sensing the above analytes. The practical application for monitoring NZF/Fe3+ in water samples was also tested.
Collapse
Affiliation(s)
- Guoxuan Xian
- Shandong Provincial Key Laboratory of Chemical Energy Storage and Novel Cell Technology, School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong Province 252059, People's Republic of China
| | - Luyao Wang
- Shandong Provincial Key Laboratory of Chemical Energy Storage and Novel Cell Technology, School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong Province 252059, People's Republic of China
| | - Xiaoyu Wan
- Shandong Provincial Key Laboratory of Chemical Energy Storage and Novel Cell Technology, School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong Province 252059, People's Republic of China
| | - Hui Yan
- School of Pharmacy, Liaocheng University, Liaocheng, Shandong Province 252059, People's Republic of China
| | - Jiawei Cheng
- Shandong Provincial Key Laboratory of Chemical Energy Storage and Novel Cell Technology, School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong Province 252059, People's Republic of China
| | - Yuqian Chen
- Shandong Provincial Key Laboratory of Chemical Energy Storage and Novel Cell Technology, School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong Province 252059, People's Republic of China
| | - Jing Lu
- Shandong Provincial Key Laboratory of Chemical Energy Storage and Novel Cell Technology, School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong Province 252059, People's Republic of China
| | - Yunwu Li
- Shandong Provincial Key Laboratory of Chemical Energy Storage and Novel Cell Technology, School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong Province 252059, People's Republic of China
| | - Dacheng Li
- Shandong Provincial Key Laboratory of Chemical Energy Storage and Novel Cell Technology, School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong Province 252059, People's Republic of China
| | - Jianmin Dou
- Shandong Provincial Key Laboratory of Chemical Energy Storage and Novel Cell Technology, School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong Province 252059, People's Republic of China
| | - Suna Wang
- Shandong Provincial Key Laboratory of Chemical Energy Storage and Novel Cell Technology, School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong Province 252059, People's Republic of China
| |
Collapse
|
35
|
Parashar P, Mazhar I, Kanoujia J, Yadav A, Kumar P, Saraf SA, Saha S. Appraisal of anti-gout potential of colchicine-loaded chitosan nanoparticle gel in uric acid-induced gout animal model. Arch Physiol Biochem 2022; 128:547-557. [PMID: 31852265 DOI: 10.1080/13813455.2019.1702702] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Present study is aimed at transdermal delivery of colchicine-loaded chitosan nanoparticles. The nanoformulations were prepared utilising spontaneous emulsification method and optimised through 23 factorial designs. The optimised formulation (CHNP-OPT) displayed an average particle size of 294 ± 3.75 nm, entrapment efficiency 92.89 ± 1.1% and drug content 83.45 ± 2.5%, respectively. In vitro release study demonstrated 89.34 ± 2.90% release over a period of 24 h. Further, CHNP-OPT incorporated into HPMC-E4M (hydroxypropyl methylcellulose) to form transdermal gel. CHNPgel displayed 74.65 ± 1.90% permeation and stability over a period of 90 days. The anti-gout potential of CHNPgel formulation was evaluated in vivo against monosodium urate (MSU) crystal-induced gout in animal model. There was significant reduction in uric acid level, during MSU administration, when compared with the conventional gel of colchicine. The enhanced therapeutic potential was witnessed through X-ray. The study revealed that colchicine-loaded CHNPgel proved their supremacy over plain colchicine and can be an efficient delivery system for gout treatment.
Collapse
Affiliation(s)
- Poonam Parashar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Ifrah Mazhar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Jovita Kanoujia
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Abhishek Yadav
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Pranesh Kumar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Shubhini A Saraf
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Sudipta Saha
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| |
Collapse
|
36
|
Tang M, Li J, Cai X, Sun T, Chen C. Single-atom Nanozymes for Biomedical Applications: Recent Advances and Challenges. Chem Asian J 2022; 17:e202101422. [PMID: 35143111 DOI: 10.1002/asia.202101422] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/08/2022] [Indexed: 11/07/2022]
Abstract
Nanozymes have received extensive attention in the fields of sensing and detection, medical therapy, industry, and agriculture thanks to the combination of the catalytic properties of natural enzymes and the physicochemical properties of nanomaterials, coupled with superior stability and ease of preparation. Despite the promise of nanozymes, conventional nanozymes are constrained by their oversized size and low catalytic capacity in sophisticated practical application environments. single-atom nanozymes (SAzymes) were characterized as nanozymes with high catalytic efficiency by uniformly distributed single atoms as catalysis sites, thus effectively addressing the defects of conventional nanozymes. This paper reviews the activity improvement scheme and catalytic mechanism of SAzymes and highlights the latest research progress of SAzymes in the fields of biomedical sensing and therapy. Eventually, the challenges and future directions of SAzymes are discussed in this paper.
Collapse
Affiliation(s)
- Minglu Tang
- Northeast Forestry University, Department of chemistry, CHINA
| | - Jingqi Li
- Northeast Forestry University, Department of chemistry, CHINA
| | - Xinda Cai
- Northeast Forestry University, Department of chemistry, CHINA
| | - Tiedong Sun
- Northeast Forestry University, 26 Hexing road, Xiangfang district, Harbin city, Heilongjiang province, 150040, Harbin, CHINA
| | - Chunxia Chen
- Northeast Forestry University, Department of chemistry, CHINA
| |
Collapse
|
37
|
Zhou GQ, Chen G, Yang J, Qin WY, Ping J. Guizhi-Shaoyao-Zhimu decoction attenuates monosodium urate crystal-induced inflammation through inactivation of NF-κB and NLRP3 inflammasome. JOURNAL OF ETHNOPHARMACOLOGY 2022; 283:114707. [PMID: 34619319 DOI: 10.1016/j.jep.2021.114707] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/18/2021] [Accepted: 10/01/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Guizhi-Shaoyao-Zhimu decoction (GSZD), a classical traditional Chinese medicine (TCM) prescription, is used empirically to treat various types of arthritis in TCM clinical practice. However, the underlying mechanisms of GSZD on gouty inflammation are not totally elucidated. AIM OF STUDY The purpose of this study is to investigate the effects of GSZD on peritoneal recruitment of neutrophils, production of proinflammatory mediators, activations of nuclear factor (NF)-κB and nucleotide oligomerization domain-like receptor protein-3 (NLRP3) inflammasome in mice with monosodium urate crystal (MSU)-induced peritonitis (MIP). MATERIALS AND METHODS Mice were intragastrically administered with GSZD for 7 days. After the last administration, mice were intraperitoneally injected with MSU. Peritoneal exudates of mice were harvested, and total peritoneal cells were calculated. Levels of interleukin (IL)-1β, IL-6 and monocyte chemotactic protein (MCP)-1 in peritoneal exudates were tested by enzyme-linked immunosorbent assay. Expressions of IL-1β, NLRP3, cysteinyl aspartate specific proteinase (caspase)-1, apoptosis-associated speck-like protein containing the caspase activation and recruitment domain (ASC), phosphorylated (p)-p65, inhibitor of NF-κB (IκB)α, p-IκB kinase (IKK)β, nuclear p65, p-mitogen-activated protein kinases (MAPKs) in peritoneal cells were analyzed by Western blot. Binding activity of NF-κB to DNA was measured by a Trans AM™ kit for p65. Interaction between ASC and pro-caspase-1 was assessed by co-immunoprecipitation assay. RESULTS Total peritoneal cells, levels of IL-1β, IL-6 and MCP-1 were significantly reduced by GSZD treatment in peritoneal exudates of MIP mice. As for the activation of NF-κB, GSZD treatment significantly reduced the levels of p-p65, p-IKKβ, nuclear p65 and p-MAPKs, enhanced the level of IκBα and abated the binding ability of NF-κB to DNA in peritoneal cells of MIP mice. As for the activation of NLRP3 inflammasome, GSZD treatment significantly reduced the levels of IL-1β, NLRP3 and caspase-1, and alleviated the interaction between ASC and pro-caspase-1 in peritoneal cells of MIP mice. Nevertheless, GSZD didn't remarkably change the level of ASC. CONCLUSIONS These results suggest that GSZD attenuates the MSU-induced inflammation through inhibiting the activations of NF-κB and NLRP3 inflammasome.
Collapse
Affiliation(s)
- Guo-Qing Zhou
- Department of Combination of Chinese and Western Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Gang Chen
- Key Laboratory of Natural Medicine Research of Chongqing Education Commission, College of Environment and Resources, Chongqing Technology and Business University, Chongqing, 400067, China.
| | - Juan Yang
- Department of Combination of Chinese and Western Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Wen-Yi Qin
- Department of Combination of Chinese and Western Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Jia Ping
- Department of Combination of Chinese and Western Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
38
|
Li H, Zhang X, Gu L, Li Q, Ju Y, Zhou X, Hu M, Li Q. Anti-Gout Effects of the Medicinal Fungus Phellinus igniarius in Hyperuricaemia and Acute Gouty Arthritis Rat Models. Front Pharmacol 2022; 12:801910. [PMID: 35087407 PMCID: PMC8787200 DOI: 10.3389/fphar.2021.801910] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/16/2021] [Indexed: 12/23/2022] Open
Abstract
Background:Phellinus igniarius (P. igniarius) is an important medicinal and edible fungus in China and other Southeast Asian countries and has diverse biological activities. This study was performed to comparatively investigate the therapeutic effects of wild and cultivated P. igniarius on hyperuricaemia and gouty arthritis in rat models. Methods: UPLC-ESI-qTOF-MS was used to identify the chemical constituents of polyphenols from wild P. igniarius (WPP) and cultivated P. igniarius (CPP). Furthermore, WPP and CPP were evaluated in an improved hyperuricaemia rat model induced by yeast extract, adenine and potassium oxonate, which was used to examine xanthine oxidase (XO) activity inhibition and anti-hyperuricemia activity. WPP and CPP therapies for acute gouty arthritis were also investigated in a monosodium urate (MSU)-induced ankle swelling model. UHPLC-QE-MS was used to explore the underlying metabolic mechanisms of P. igniarius in the treatment of gout. Results: The main active components of WPP and CPP included protocatechuic aldehyde, hispidin, davallialactone, phelligridimer A, hypholomine B and inoscavin A as identified by UPLC-ESI-qTOF-MS. Wild P. igniarius and cultivated P. igniarius showed similar activities in reducing uric acid levels through inhibiting XO activity and down-regulating the levels of UA, Cr and UN, and they had anti-inflammatory activities through down-regulating the secretions of ICAM-1, IL-1β and IL-6 in the hyperuricaemia rat model. The pathological progression of kidney damage was also reversed. The polyphenols from wild and cultivated P. igniarius also showed significant anti-inflammatory activity by suppressing the expression of ICAM-1, IL-1β and IL-6 and by reducing the ankle joint swelling degree in an MSU-induced acute gouty arthritis rat model. The results of metabolic pathway enrichment indicated that the anti-hyperuricemia effect of WPP was mainly related to the metabolic pathways of valine, leucine and isoleucine biosynthesis and histidine metabolism. Additionally, the anti-hyperuricemia effect of CPP was mainly related to nicotinate and nicotinamide metabolism and beta-alanine metabolism. Conclusions: Wild P. igniarius and cultivated P. igniarius both significantly affected the treatment of hyperuricaemia and acute gouty arthritis models in vivo and therefore may be used as potential active agents for the treatment of hyperuricaemia and acute gouty arthritis.
Collapse
Affiliation(s)
- Hongxing Li
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China.,Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Xinyue Zhang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China.,Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Lili Gu
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China.,Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Qín Li
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China.,Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Yue Ju
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China.,Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Xuebin Zhou
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China.,Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Min Hu
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China.,Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Qīn Li
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China.,Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
39
|
Wu Y, Ye Z, Feng P, Li R, Chen X, Tian X, Han R, Kakade A, Liu P, Li X. Limosilactobacillus fermentum JL-3 isolated from "Jiangshui" ameliorates hyperuricemia by degrading uric acid. Gut Microbes 2022; 13:1-18. [PMID: 33764849 PMCID: PMC8007157 DOI: 10.1080/19490976.2021.1897211] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Recent studies into the beneficial effects of fermented foods have shown that this class of foods are effective in managing hyperuricemia and gout. In this study, the uric acid (UA) degradation ability of Limosilactobacillus fermentum JL-3 strain, isolated from "Jiangshui" (a fermented Chinese food), was investigated. In vitro results showed that JL-3 strain exhibited high degradation capacity and selectivity toward UA. After oral administration to mice for 15 days, JL-3 colonization was continuously detected in the feces of mice. The UA level in urine of mice fed with JL-3 was similar with the control group mice. And the serum UA level of the former was significantly lower (31.3%) than in the control, further confirmed the UA-lowering effect of JL-3 strain. Limosilactobacillus fermentum JL-3 strain also restored some of the inflammatory markers and oxidative stress indicators (IL-1β, MDA, CRE, blood urea nitrogen) related to hyperuricemia, while the gut microbial diversity results showed that JL-3 could regulate gut microbiota dysbiosis caused by hyperuricemia. Therefore, the probiotic Limosilactobacillus fermentum JL-3 strain is effective in lowering UA levels in mice and could be used as a therapeutic adjunct agent in treating hyperuricemia.
Collapse
Affiliation(s)
- Ying Wu
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, China,Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, China
| | - Ze Ye
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Pengya Feng
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, China
| | - Rong Li
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, China,Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, China
| | - Xiao Chen
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, China
| | - Xiaozhu Tian
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, China,Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, China
| | - Rong Han
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, China
| | - Apurva Kakade
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, China
| | - Pu Liu
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, China,CONTACT Xiangkai Li Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, China
| | - Xiangkai Li
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, China,Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, China
| |
Collapse
|
40
|
Wang J, Liu K, Xiao T, Liu P, Prinz RA, Xu X. Uric acid accumulation in DNA-damaged tumor cells induces NKG2D ligand expression and antitumor immunity by activating TGF-β-activated kinase 1. Oncoimmunology 2022; 11:2016159. [PMID: 35154904 PMCID: PMC8837239 DOI: 10.1080/2162402x.2021.2016159] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
DNA damage by genotoxic drugs such as gemcitabine and 5-fluorouracil (5-FU) activates the ataxia telangiectasia, mutated (ATM)-Chk pathway and induces the expression of NKG2D ligands such as the MHC class I-related chain A and B (MICA/B). The mechanisms underlying this remain incompletely understood. Here we report that xanthine oxidoreductase (XOR), a rate-limiting enzyme that produces uric acid in the purine catabolism pathway, promotes DNA damage-induced MICA/B expression. Inhibition of the ATM-Chk pathway blocks genotoxic drug-induced uric acid production, TGF-β-activated kinase 1 (TAK1) activation, ERK phosphorylation, and MICA/B expression. Inhibition of uric acid production by the XOR inhibitor allopurinol blocks DNA damage-induced TAK1 activation and MICA/B expression in genotoxic drug-treated cells. Exogenous uric acid activates TAK1, NF-κB, and the MAP kinase pathway. TAK1 inhibition blocks gemcitabine- and uric acid-induced MAP kinase activation and MICA/B expression. Exogenous uric acid in its salt form, monosodium urate (MSU), induces MICA/B expression and sensitizes tumor cells to NK cell killing. MSU immunization with irradiated murine breast cancer cell line RCAS-Neu retards breast cancer growth in syngeneic breast cancer models and delays breast cancer development in a somatic breast cancer model. Our study suggests that uric acid accumulation plays an important role in activating TAK1, inducing DNA damage-induced MICA/B expression, and enhancing antitumor immunity.
Collapse
Affiliation(s)
- Jingxiang Wang
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Kai Liu
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Tianxiang Xiao
- College of Medicine, Yangzhou University, Yangzhou Jiangsu Province, P. R. China
| | - Penggang Liu
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Richard A. Prinz
- Department of Surgery, NorthShore University Health System, Evanston, IL USA
| | - Xiulong Xu
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, Jiangsu Province, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou Jiangsu Province, China
- Department of Molecular and Cellular Medicine, Rush University Medical Center, Chicago, Il USA
| |
Collapse
|
41
|
Chen G, Guo T, Yang L. Paeonol reduces IL-β production by inhibiting the activation of nucleotide oligomerization domain-like receptor protein-3 inflammasome and nuclear factor-κB in macrophages. Biochem Cell Biol 2021; 100:28-36. [PMID: 34784237 DOI: 10.1139/bcb-2021-0255] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Interleukin-1β, a key cytokine in gouty inflammation, is precisely regulated by the NLRP3 inflammasome and NF-κB. Our previous study demonstrated that paeonol suppressed IL-1β production in rats with monosodium urate (MSU)-induced arthritis. Whether NLRP3 inflammasome or NF-κB is responsible for the anti-inflammatory effect of paeonol remains unclear. In this study, J774A.1 cells induced by lipopolysaccharide (LPS) plus MSU, was used to investigate the effect of paeonol on NLRP3 inflammasome activation, and J774A.1 cells induced by LPS alone were used to investigate the effect of paeonol on NF-κB activation. In J774A.1 cells induced by LPS plus MSU, paeonol decreased the levels of IL-1β and caspase-1 and reduced the MSU-induced interaction of pro-caspase-1 and apoptosis-associated speck-like protein containing caspase recruitment domain (ASC), but did not affect the levels of pro-IL-1β and pro-caspase-1. In J774A.1 cells induced by LPS alone, paeonol reduced the levels of IL-1β, NLRP3, p-IKK, p-IκBα, and p-p65, but did not affect ASC levels. Paeonol also promoted the content of IκBα and retained more p65 in the cytoplasm. Furthermore, paeonol reduced the DNA-binding activity of p65 and lowered the levels of p-JNK, p-ERK, and p-p38. These results suggest that paeonol inhibits IL-1β production by inhibiting the activation of NLRP3 inflammasome, NF-κB, and MAPK signaling pathways.
Collapse
Affiliation(s)
- Gang Chen
- Key Laboratory of Natural Medicine Research of Chongqing Education Commission, College of Environment and Resources, Chongqing Technology and Business University, Chongqing 400067, China.,Key Laboratory of Natural Medicine Research of Chongqing Education Commission, College of Environment and Resources, Chongqing Technology and Business University, Chongqing 400067, China
| | - Tingwang Guo
- Key Laboratory of Natural Medicine Research of Chongqing Education Commission, College of Environment and Resources, Chongqing Technology and Business University, Chongqing 400067, China.,Key Laboratory of Natural Medicine Research of Chongqing Education Commission, College of Environment and Resources, Chongqing Technology and Business University, Chongqing 400067, China
| | - Lin Yang
- Key Laboratory of Natural Medicine Research of Chongqing Education Commission, College of Environment and Resources, Chongqing Technology and Business University, Chongqing 400067, China.,Key Laboratory of Natural Medicine Research of Chongqing Education Commission, College of Environment and Resources, Chongqing Technology and Business University, Chongqing 400067, China
| |
Collapse
|
42
|
Kim SK, Park KY, Choe JY. Toll-Like Receptor 9 Is Involved in NLRP3 Inflammasome Activation and IL-1β Production Through Monosodium Urate-Induced Mitochondrial DNA. Inflammation 2021; 43:2301-2311. [PMID: 32700178 DOI: 10.1007/s10753-020-01299-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The NLR family pyrin domain-containing 3 (NLRP3) inflammasome is a cytoplasmic multimolecular complex that generates interleukin (IL)-1β and is considered a main pathogenic mechanism for uric acid-induced inflammation. Whether toll-like receptor 9 (TLR9) is responsible for uric acid-induced NLRP3 inflammasome activation remains unclear. Thus, the aim of this study was to identify the role of TLR9 in NLRP3 inflammasome activation through monosodium urate (MSU) crystal-induced mitochondrial DNA. RAW 264.7 cells treated with MSU crystals, CpG oligonucleotides (ODNs), or a combination of both were used to assess nuclear factor (NF)-κB signaling, NLRP3 inflammasome components such as NLRP3, ASC, and caspase-1, and IL-1β. Real-time polymerase chain reaction (RT-PCR), Western blotting, DNA fragmentation assay, mitochondrial DNA copy number assay, and immunofluorescence were used in the in vitro study. RAW 264.7 cells treated with CpG-ODN stimulated the activation of NF-κB signaling, the NLRP3 inflammasome components NLRP3, ASC, and caspase-1, and IL-1β gene and protein expression. DNA fragmentation assay showed that MSU crystals induced cellular apoptosis. Fragmented DNA prompted by MSU crystals induced TLR9 expression. RAW 264.7 cells treated with CpG-ODN or MSU crystals and both increased expression of mitochondrial DNA relative to nuclear DNA. CpG-ODN and MSU crystals augmented the activation of NLRP3 inflammasome components and IL-1β expression, which was significantly suppressed in RAW 264.7 cells transfected with TLR9 siRNA. This study suggests that TLR9 activated by MSU crystal-mediated mitochondrial DNA contributes to the activation of NLRP3 inflammasomes and IL-1β production.
Collapse
Affiliation(s)
- Seong-Kyu Kim
- Department of Internal Medicine, Division of Rheumatology, Catholic University of Daegu School of Medicine, Daegu, Republic of Korea. .,Arthritis and Autoimmunity Research Center, Catholic University of Daegu, Daegu, Republic of Korea.
| | - Ki-Yeun Park
- Arthritis and Autoimmunity Research Center, Catholic University of Daegu, Daegu, Republic of Korea
| | - Jung-Yoon Choe
- Department of Internal Medicine, Division of Rheumatology, Catholic University of Daegu School of Medicine, Daegu, Republic of Korea.,Arthritis and Autoimmunity Research Center, Catholic University of Daegu, Daegu, Republic of Korea
| |
Collapse
|
43
|
王 贵, 左 婷, 李 然, 左 正. [Effect of rebamipide on the acute gouty arthritis in rats induced by monosodium urate crystals]. BEIJING DA XUE XUE BAO. YI XUE BAN = JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2021; 53:716-720. [PMID: 34393234 PMCID: PMC8365083 DOI: 10.19723/j.issn.1671-167x.2021.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Indexed: 06/13/2023]
Abstract
OBJECTIVE To investigate the role of rebamipide in the treatment of acute gout arthritis rats induced by monosodium urate (MSU) crystal. METHODS Forty-two male rats were randomly divided into three groups (n=14). Group A was treated with oral rebamipide, group B with oral colchicine, and group C with oral placebo. The rats were monitored for the induction of arthritis with clinical manifestations and pathological changes, and the levels of interleukin (IL)-1β、IL-6、IL-10, and tumor necrosis factor (TNF)-α in serum were measured. RESULTS In group C, the clinical score and swelling index reached the maximum in 24 h, and then gradually decreased to 72 h. After 24 h of model induced, the clinical scores in group C were significantly higher than those in group A and group B [2 (1-3) vs. 0 (0-1) vs. 1 (0-2), P < 0.01], the swelling indexes in group C were significantly higher than those in group A and group B [0.36 (0.16-0.52) vs. 0.11 (0-0.20) vs. 0.12 (0-0.16), P < 0.01]. Histologically, after 24 h of model induced, there was a large number of neutrophil infiltration in the synovium of group C [scale score: 4 (2-4)], and there was no significant inflammatory cell infiltration in group A [1 (0-2)] and group B [1 (0-2)], the difference was statistically significant (P < 0.001). After 24 h of model induced, the levels of IL-1β, IL-6, IL-10, and TNF-α in serum of group C were significantly higher than those in group A and B [IL-1β: (41.86±5.72) vs. (27.35±7.47) vs. (27.76±5.28) ng/L, IL-6: (1 575.55±167.11) vs. (963.53±90.22) vs. (964.08±99.31) ng/L, IL-10: (37.96±3.76) vs. (21.68±4.83) vs. (16.20±2.49) ng/L, TNF-α: (21.32±1.34) vs. (15.82±2.54) vs. (17.35±7.47) μg/L, P < 0.001]. CONCLUSION Rebamipide has a protective effect on acute gout arthritis rats induced by MUS crystals.
Collapse
Affiliation(s)
- 贵红 王
- />安徽医科大学附属安庆医院风湿免疫科,安徽安庆 246000Department of Rheumatology and Immunology, Anqing Hospital of Anhui Medical University, Anqing 246000, Anhui, China
| | - 婷 左
- />安徽医科大学附属安庆医院风湿免疫科,安徽安庆 246000Department of Rheumatology and Immunology, Anqing Hospital of Anhui Medical University, Anqing 246000, Anhui, China
| | - 然 李
- />安徽医科大学附属安庆医院风湿免疫科,安徽安庆 246000Department of Rheumatology and Immunology, Anqing Hospital of Anhui Medical University, Anqing 246000, Anhui, China
| | - 正才 左
- />安徽医科大学附属安庆医院风湿免疫科,安徽安庆 246000Department of Rheumatology and Immunology, Anqing Hospital of Anhui Medical University, Anqing 246000, Anhui, China
| |
Collapse
|
44
|
Kim ST, Tayar J, Fu S, Ke D, Norry E, Sun A, Miller J, Hong DS. Newly developed pseudogout arthritis after therapy with MAGE-A4 directed TCR T cells responded to treatment with tocilizumab. J Immunother Cancer 2021; 9:jitc-2021-002716. [PMID: 34233963 PMCID: PMC8264871 DOI: 10.1136/jitc-2021-002716] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2021] [Indexed: 11/04/2022] Open
Abstract
With durable cancer responses, genetically modified cell therapies are being implemented in various cancers. However, these immune effector cell therapies can cause toxicities, including cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Pseudogout arthritis is an inflammatory arthritis induced by deposition of calcium pyrophosphate dihydrate crystals. Here, we report a case of pseudogout arthritis in a patient treated with MAGE-A4 directed T cell receptor T cells, for fallopian tube cancer. The patient developed CRS and ICANS 7 days after infusion of the T cells. Concurrently, the patient newly developed sudden onset of left knee arthritis. Synovial fluid analyses revealed the presence of calcium pyrophosphate dihydrate crystal. Notably, the pseudogout arthritis was resolved with tocilizumab, which was administered for the treatment of CRS and ICANS. Immunoprofiling of the synovial fluid showed that the proportion of inflammatory interleukin 17 (IL-17)-producing CD4+ T (Th17) cells and amount of IL-6 were notably increased, suggesting a potential role of Th17 cells in pseudogout arthritis after T-cell therapy. To the best of our knowledge, this is the first reported case of pseudogout arthritis after cell therapy. Clinicians, especially hematologists, oncologists and rheumatologists, should be aware that pseudogout arthritis can be associated with CRS/ICANS.
Collapse
Affiliation(s)
- Sang T Kim
- General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jean Tayar
- General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Siqing Fu
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Danxia Ke
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Elliot Norry
- Adaptimmune Therapeutics plc, Philadelphia, Pennsylvania, USA
| | - Amy Sun
- Adaptimmune Therapeutics plc, Philadelphia, Pennsylvania, USA
| | - Juli Miller
- Adaptimmune Therapeutics plc, Philadelphia, Pennsylvania, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
45
|
Li WY, Yang F, Chen JH, Ren GF. β-Caryophyllene Ameliorates MSU-Induced Gouty Arthritis and Inflammation Through Inhibiting NLRP3 and NF-κB Signal Pathway: In Silico and In Vivo. Front Pharmacol 2021; 12:651305. [PMID: 33967792 PMCID: PMC8103215 DOI: 10.3389/fphar.2021.651305] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 04/12/2021] [Indexed: 01/17/2023] Open
Abstract
Gouty arthritis serves as an acute reaction initiated by the deposition of monosodium urate (MSU) crystals around the joints. In this study, the anti-inflammatory effects of phytochemical β-caryophyllene on MSU crystal-induced acute gouty arthritis in vivo and in silico were explored. Through bioinformatics methods and molecular docking, it screened the specific influence pathway of β-caryophyllene on gout. Certain methods including enzyme-linked immunosorbent assay, western blotting, and immunohistochemical staining were adopted to quantify. β-caryophyllene significantly reduced inflammation and function of ankle joints in MSU Crystals-induced gouty arthritis rats, while decreasing serum cytokine levels. Furthermore, it inhibited the expressions of NLRP3, Caspase-1, ASC, TLR4, MyD88, p65, and IL-1β in the synovial tissue so as to reduce inflammation and protect ankle joints’ function. A new research approach in which β-caryophyllene treatment to acute attacks of gout is provided through the research results.
Collapse
Affiliation(s)
- Wan-Yang Li
- Xiangya School of Public Health, Central South University, Changsha, China
| | - Fan Yang
- School of Chinese Traditional Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ji-Hua Chen
- Xiangya School of Public Health, Central South University, Changsha, China
| | - Guo-Feng Ren
- Xiangya School of Public Health, Central South University, Changsha, China
| |
Collapse
|
46
|
Li H, Uittenbogaard M, Hao L, Chiaramello A. Clinical Insights into Mitochondrial Neurodevelopmental and Neurodegenerative Disorders: Their Biosignatures from Mass Spectrometry-Based Metabolomics. Metabolites 2021; 11:233. [PMID: 33920115 PMCID: PMC8070181 DOI: 10.3390/metabo11040233] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 04/01/2021] [Accepted: 04/07/2021] [Indexed: 02/07/2023] Open
Abstract
Mitochondria are dynamic multitask organelles that function as hubs for many metabolic pathways. They produce most ATP via the oxidative phosphorylation pathway, a critical pathway that the brain relies on its energy need associated with its numerous functions, such as synaptic homeostasis and plasticity. Therefore, mitochondrial dysfunction is a prevalent pathological hallmark of many neurodevelopmental and neurodegenerative disorders resulting in altered neurometabolic coupling. With the advent of mass spectrometry (MS) technology, MS-based metabolomics provides an emerging mechanistic understanding of their global and dynamic metabolic signatures. In this review, we discuss the pathogenetic causes of mitochondrial metabolic disorders and the recent MS-based metabolomic advances on their metabolomic remodeling. We conclude by exploring the MS-based metabolomic functional insights into their biosignatures to improve diagnostic platforms, stratify patients, and design novel targeted therapeutic strategies.
Collapse
Affiliation(s)
- Haorong Li
- Department of Chemistry, George Washington University, Science and Engineering Hall 4000, 800 22nd St., NW, Washington, DC 20052, USA;
| | - Martine Uittenbogaard
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, George Washington University, 2300 I Street N.W. Ross Hall 111, Washington, DC 20037, USA;
| | - Ling Hao
- Department of Chemistry, George Washington University, Science and Engineering Hall 4000, 800 22nd St., NW, Washington, DC 20052, USA;
| | - Anne Chiaramello
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, George Washington University, 2300 I Street N.W. Ross Hall 111, Washington, DC 20037, USA;
| |
Collapse
|
47
|
Valsalan Soba S, Babu M, Panonnummal R. Ethosomal Gel Formulation of Alpha Phellandrene for the Transdermal Delivery in Gout. Adv Pharm Bull 2021; 11:137-149. [PMID: 33747861 PMCID: PMC7961230 DOI: 10.34172/apb.2021.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 02/24/2020] [Accepted: 04/16/2020] [Indexed: 12/29/2022] Open
Abstract
Purpose: Purpose was to improve the skin compatibility and permeability of alpha phellandrene through an ethosomal gel formulation for the treatment of gout; as the oral use of the drug is reported to cause gastrointestinal disturbances and toxicities. Methods: Alpha phellandrene loaded ethosomal formulation (APES) was prepared by cold method for the treatment of gout. APES were loaded into carbopol gel (APEG) by dispersion method. Physico-chemical characterizations of the APES were done by dynamic light scattering (DLS), transmission electron microscopy (TEM), Fourier-transform infrared spectroscopy (FTIR) etc. In vitro release, permeation, haemo-compatibility and anti-inflammatory studies were conducted. Results: APES showed a particle size of 364.83 ± 45.84 nm. The entrapment efficiency of the optimized formulation is found as 95.06 ± 2.51%. Hemolysis data indicated that APES does not cause any significant hemolysis. In vitro drug release studies were carried out using dialysis membrane technique and the amount of drug released from APES & APEG is found to be 95% and 94.21% respectively after 5 and6 hours. Kinetic data analysis revealed that APES & APEG follows first order and zero order release kinetics, respectively. The anti-inflammatory activity studies of the formulation are done by estimating its inhibitory effects on cyclooxygenase II (COX) II, lipoxygenase-5 (LOX-5), Myeloperoxidase (MPO), Inducible nitric oxide synthase (INOS) & cellular nitrite level using RAW 264.7 cells. The significant inhibition in the activities of the enzymes implies the anti-inflammatory activity of the formulations. Skin permeation study was carried out using porcine skin and revealed that the permeation of alpha phellandrene is increased from APES & APEG when compared with alpha-phellandrene solution (APS). Skin deposition study of APS, APES & APEG revealed better drug deposition from APEG (48.799 ± 1.547µg/cm2 ) after 24 hours when compared with APS & APES. Conclusion: Overall results indicate that the ethosomal formulation of alpha phellandrene through transdermal route is an effective alternative for oral use of the drug.
Collapse
Affiliation(s)
- Sony Valsalan Soba
- Amrita School of Pharmacy, Amrita Institute of Medical Science & Research Center, Amrita VishwaVidyapeetham, Kochi-682041 India
| | - Merin Babu
- Amrita School of Pharmacy, Amrita Institute of Medical Science & Research Center, Amrita VishwaVidyapeetham, Kochi-682041 India
| | - Rajitha Panonnummal
- Amrita School of Pharmacy, Amrita Institute of Medical Science & Research Center, Amrita VishwaVidyapeetham, Kochi-682041 India
| |
Collapse
|
48
|
Hu FX, Hu T, Chen S, Wang D, Rao Q, Liu Y, Dai F, Guo C, Yang HB, Li CM. Single-Atom Cobalt-Based Electrochemical Biomimetic Uric Acid Sensor with Wide Linear Range and Ultralow Detection Limit. NANO-MICRO LETTERS 2020; 13:7. [PMID: 34138193 PMCID: PMC8187548 DOI: 10.1007/s40820-020-00536-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 09/13/2020] [Indexed: 05/17/2023]
Abstract
Uric acid (UA) detection is essential in diagnosis of arthritis, preeclampsia, renal disorder, and cardiovascular diseases, but it is very challenging to realize the required wide detection range and low detection limit. We present here a single-atom catalyst consisting of Co(II) atoms coordinated by an average of 3.4 N atoms on an N-doped graphene matrix (A-Co-NG) to build an electrochemical biomimetic sensor for UA detection. The A-Co-NG sensor achieves a wide detection range over 0.4-41,950 μM and an extremely low detection limit of 33.3 ± 0.024 nM, which are much better than previously reported sensors based on various nanostructured materials. Besides, the A-Co-NG sensor also demonstrates its accurate serum diagnosis for UA for its practical application. Combination of experimental and theoretical calculation discovers that the catalytic process of the A-Co-NG toward UA starts from the oxidation of Co species to form a Co3+-OH-UA*, followed by the generation of Co3+-OH + *UA_H, eventually leading to N-H bond dissociation for the formation of oxidized UA molecule and reduction of oxidized Co3+ to Co2+ for the regenerated A-Co-NG. This work provides a promising material to realize UA detection with wide detection range and low detection limit to meet the practical diagnosis requirements, and the proposed sensing mechanism sheds light on fundamental insights for guiding exploration of other biosensing processes.
Collapse
Affiliation(s)
- Fang Xin Hu
- Institute of Materials Science and Devices, Suzhou University of Science and Technology, Suzhou, 215009, People's Republic of China
| | - Tao Hu
- Institute of Materials Science and Devices, Suzhou University of Science and Technology, Suzhou, 215009, People's Republic of China
| | - Shihong Chen
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, People's Republic of China
| | - Dongping Wang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, People's Republic of China
| | - Qianghai Rao
- Institute of Materials Science and Devices, Suzhou University of Science and Technology, Suzhou, 215009, People's Republic of China
| | - Yuhang Liu
- Institute of Materials Science and Devices, Suzhou University of Science and Technology, Suzhou, 215009, People's Republic of China
| | - Fangyin Dai
- State Key Laboratory of Silkworm Genome Biology, College of Biotechnology, Southwest University, Chongqing, 400715, People's Republic of China
| | - Chunxian Guo
- Institute of Materials Science and Devices, Suzhou University of Science and Technology, Suzhou, 215009, People's Republic of China.
| | - Hong Bin Yang
- Institute of Materials Science and Devices, Suzhou University of Science and Technology, Suzhou, 215009, People's Republic of China.
| | - Chang Ming Li
- Institute of Materials Science and Devices, Suzhou University of Science and Technology, Suzhou, 215009, People's Republic of China.
- Institute for Advanced Cross-field Science and College of Life Science, Qingdao University, Qingdao, 200671, People's Republic of China.
- Institute for Clean Energy and Advanced Materials, School of Materials and Energy, Southwest University, Chongqing, 400715, People's Republic of China.
| |
Collapse
|
49
|
Jena M, Tripathy A, Mishra A, Maiti R. Effect of canakinumab on clinical and biochemical parameters in acute gouty arthritis: a meta-analysis. Inflammopharmacology 2020; 29:35-47. [PMID: 32918702 DOI: 10.1007/s10787-020-00753-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 08/28/2020] [Indexed: 11/27/2022]
Abstract
BACKGROUND Targeted anti-IL-1β therapy may be a valuable option for the management of gouty arthritis. The present meta-analysis has evaluated the effect of canakinumab, an anti-IL-1β monoclonal antibody in gouty arthritis. METHODS A standard meta-analysis protocol was developed and after performing a comprehensive literature search in MEDLINE, Cochrane, and International Clinical Trial Registry Platform (ICTRP), reviewers assessed eligibility and extracted data from three relevant articles. A random-effects model was used to estimate the pooled effect size as the mean difference in Visual Analouge Scale (VAS) score, serum hsCRP, serum Amyloid A, and risk ratio for global assessment between the groups. Quality assessment was done using the risk of bias assessment tool and summary of findings was prepared using standard Cochrane methodology with GradePro GDT. RESULTS Treatment with canakinumab showed a mean reduction of VAS score by 14.59 mm [95% CI - 19.42 to - 9.77], serum hsCRP by 15.36 mg/L [95% CI 1.62-29.11], serum Amyloid A by 67.18 mg/L [95% CI 17.06-117.31], and improvement in patient global assessment (RR = 1.478; 95% CI 1.29-1.67) and physician global assessment (RR = 1.44; 95% CI 1.28-1.61). The probability that future studies may have a mean difference in VAS score less than zero has been calculated to be 27.3% using a cumulative distribution function (CDF) calculator. CONCLUSION This meta-analysis shows the beneficial effect of canakinumab over triamcinolone by reducing VAS score, serum hsCRP, serum amyloid A, and improvement in global assessments in acute gouty arthritis.
Collapse
Affiliation(s)
- Monalisa Jena
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, 751019, Odisha, India
| | - Amruta Tripathy
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, 751019, Odisha, India
| | - Archana Mishra
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Rituparna Maiti
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, 751019, Odisha, India.
| |
Collapse
|
50
|
New variants in NLRP3 inflammasome genes increase risk for asthma and Blomia tropicalis-induced allergy in a Brazilian population. Cytokine X 2020; 2:100032. [PMID: 33015616 PMCID: PMC7522708 DOI: 10.1016/j.cytox.2020.100032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 06/08/2020] [Accepted: 06/15/2020] [Indexed: 02/07/2023] Open
Abstract
Atopic asthma is a chronic lung disease of lower airways caused mainly due to action of T-helper (Th) 2 type cytokines, eosinophilic inflammation, mucus hypersecretion and airway remodelling. Interleukin (IL)-33 increases type 2 immunity polarization in airway playing critical role in eosinophilic asthma. On the other hand, NLRP3 inflammasome activation results in the release of caspase-1 (Casp-1) which, in its turn, promotes IL-33 inactivation. Recent studies have shown associations between NLRP3 variants and inflammatory diseases. However, no study with genes in NLRP3 inflammassome route has been conducted so far with asthma and atopy in any population to date. Blood samples were collected from 1246 asthmatic and non-asthmatic children. Associations were tested for single nucleotide polymorphism (SNP)s in NLRP3 and CASP1 with asthma and markers of atopy and in cultures stimulated with Blomia tropicalis (Bt) mite crude extract. The T allele of rs4925648 (NLRP3) was associated with increased asthma risk (OR 1.50, P = 0.005). In addition, the T allele of rs12130711 polymorphism, whithin the same gene, acted as a protector factor for asthma (OR 0.78, P = 0.038). On the other hand, the C allele of rs4378247 NLRP3 variant was associated with lower levels of IL-13 production when peripheral blood cells were stimulated with Bt (OR 0.39, P = 4E-04). In addition, the greater the number of risk alleles in IL33/NLRP3/CASP1 route the greater was the risk for asthma. The T allele of rs7925706 CASP1 variant was also associated with increased risk for asthma (OR 1.47, P = 0.008). In addition, this same allele increased the eosinophil counts in blood (mm3) in asthmatic individuals compared with non-asthmatic (P = 0.0004). These results suggest that NLRP3 and CASP1 polymorphisms may be associated with susceptibility for asthma and markers of atopy in our population.
Collapse
|