1
|
Dabravolski SA, Churov AV, Beloyartsev DF, Kovyanova TI, Lyapina IN, Sukhorukov VN, Orekhov AN. The role of NRF2 function and regulation in atherosclerosis: an update. Mol Cell Biochem 2025:10.1007/s11010-025-05233-y. [PMID: 40025257 DOI: 10.1007/s11010-025-05233-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 02/14/2025] [Indexed: 03/04/2025]
Abstract
Atherosclerosis, a chronic inflammatory disease of the arteries, remains a leading cause of cardiovascular morbidity and mortality worldwide. This review examines the molecular mechanisms underlying NRF2 role in atherosclerosis, focusing on the recently defined intricate interplay between autophagy, the nuclear factor erythroid 2-related factor 2 (NRF2) pathway, microRNAs (miRNAs), and genes regulating NRF2 with atheroprotective effects. The NRF2/autophagy axis emerges as a critical regulator of cellular responses to oxidative stress and inflammation in atherosclerosis, with key players including Heat Shock Protein 90 (HSP90), Neuropeptide Y (NPY), and Glutaredoxin 2 (GLRX2). MiRNAs are identified as potent regulators of gene expression in atherosclerosis, impacting NRF2 signalling and disease susceptibility. Additionally, genes such as Prenyl diphosphate synthase subunit 2 (PDSS2), Sulfiredoxin1 (Srxn1), and Isocitrate dehydrogenase 1 (IDH1) are implicated in NRF2-dependent atheroprotective pathways. Future research directions include elucidating the complex interactions between these molecular pathways, evaluating novel therapeutic targets in preclinical and clinical settings, and addressing challenges related to drug delivery and patient heterogeneity. Despite limitations, this review underscores the potential for targeted interventions aimed at modulating NRF2/autophagy signalling and miRNA regulatory networks to mitigate atherosclerosis progression and improve cardiovascular outcomes.
Collapse
Affiliation(s)
- Siarhei A Dabravolski
- Department of Biotechnology Engineering, Braude Academic College of Engineering, Snunit 51, P.O. Box 78, 2161002, Karmiel, Israel.
| | - Alexey V Churov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, Russia, 125315
| | - Dmitry F Beloyartsev
- Vascular Surgery Department, A. V. Vishnevsky National Medical Research Center of Surgery, 27 Bolshaya Serpukhovskaya Street, Moscow, Russia, 117997
| | - Tatiana I Kovyanova
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, Russia, 125315
| | - Irina N Lyapina
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Barbarash Boulevard, Kemerovo, Russia, 650002
| | - Vasily N Sukhorukov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, Russia, 125315
| | - Alexander N Orekhov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, Russia, 125315
| |
Collapse
|
2
|
Zhu B, Huang X, Zhang J, Wang X, Tian S, Zhan T, Liu Y, Zhang H, Chen S, Yu C. A New Perspective on the Prediction and Treatment of Stroke: The Role of Uric Acid. Neurosci Bull 2025; 41:486-500. [PMID: 39312108 PMCID: PMC11876515 DOI: 10.1007/s12264-024-01301-3] [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: 02/08/2024] [Accepted: 07/28/2024] [Indexed: 03/04/2025] Open
Abstract
Stroke, a major cerebrovascular disease, has high morbidity and mortality. Effective methods to reduce the risk and improve the prognosis are lacking. Currently, uric acid (UA) is associated with the pathological mechanism, prognosis, and therapy of stroke. UA plays pro/anti-oxidative and pro-inflammatory roles in vivo. The specific role of UA in stroke, which may have both neuroprotective and damaging effects, remains unclear. There is a U-shaped association between serum uric acid (SUA) levels and ischemic stroke (IS). UA therapy provides neuroprotection during reperfusion therapy for acute ischemic stroke (AIS). Urate-lowering therapy (ULT) plays a protective role in IS with hyperuricemia or gout. SUA levels are associated with the cerebrovascular injury mechanism, risk, and outcomes of hemorrhagic stroke. In this review, we summarize the current research on the role of UA in stroke, providing potential targets for its prediction and treatment.
Collapse
Affiliation(s)
- Bingrui Zhu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
| | - Xiaobin Huang
- Department of Neurosurgery, The Second People's Hospital of Quzhou, Quzhou, 324000, China
| | - Jiahao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
| | - Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
| | - Sixuan Tian
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
| | - Tiantong Zhan
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
| | - Yibo Liu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
| | - Haocheng Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
| | - Sheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China.
| | - Cheng Yu
- Department of Neurosurgery, The Second People's Hospital of Quzhou, Quzhou, 324000, China.
| |
Collapse
|
3
|
Dogru S, Yasar E, Yesilkaya A. Effects of uric acid on oxidative stress in vascular smooth muscle cells. Biomed Rep 2024; 21:171. [PMID: 39346579 PMCID: PMC11428086 DOI: 10.3892/br.2024.1859] [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: 04/29/2024] [Accepted: 08/15/2024] [Indexed: 10/01/2024] Open
Abstract
Hyperuricemia during hypertension is associated with aberrant vascular functions and increased oxidative stress, which affects endothelial dysfunction. Nevertheless, the molecular mechanisms underlying the effects of uric acid on vascular smooth muscle cells (VSMCs) through oxidative stress remain unclear. The aim of the present study was to investigate the dose- and time-dependent effects of uric acid on oxidative stress and p53 protein expression in VSMCs. VSMCs were incubated with various concentrations of uric acid (0-50 mg/dl) for different time periods (1-24 h). Thiobarbituric acid reactive substances (TBARs), protein carbonylation and nitric oxide (NO) levels were determined using appropriate assay kits. Superoxide anion release was detected using the Görlach method. Western blotting was performed to determine the protein expression levels of p53. The findings demonstrated that the application of uric acid led to an increase in protein carbonylation and superoxide anion levels while causing a decrease in NO levels. Conversely, no significant effect was observed on TBARS levels. Additionally, it was observed that high concentrations of uric acid suppressed p53 expression at 6, 12 and 24 h. The present study provided evidence that the influence of uric acid on oxidative stress was more closely associated with time than dose; however, not all effects observed were strictly time-dependent.
Collapse
Affiliation(s)
- Segun Dogru
- Department of Medical Biochemistry, Akdeniz University Medical School, 07058 Antalya, Turkey
| | - Ekrem Yasar
- Department of Biophysics, Akdeniz University Medical School, 07058 Antalya, Turkey
- Department of Biophysics, Faculty of Medicine, Erzincan Binali Yildirim University, 24100 Erzincan, Turkey
| | - Akin Yesilkaya
- Department of Medical Biochemistry, Akdeniz University Medical School, 07058 Antalya, Turkey
| |
Collapse
|
4
|
Xie JB, Huang SJ, Yang TB, Wang W, Chen BY, Guo L. Comparison of machine learning methods for Predicting 3-Year survival in elderly esophageal squamous cancer patients based on oxidative stress. BMC Cancer 2024; 24:1432. [PMID: 39574068 PMCID: PMC11580478 DOI: 10.1186/s12885-024-13115-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/28/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Oxidative stress process plays a key role in aging and cancer; however, currently, there is paucity of machine-learning model studies investigating the relationship between oxidative stress and prognosis of elderly patients with esophageal squamous cancer (ESCC). METHODS This study included elderly patients with ESCC who underwent curative ESCC resection surgery continuously from January 2013 to December 2020 and were stratified into the training and external validation cohorts. Using Cox stepwise regression analysis based on Akaike information criterion, the relationship between oxidative stress biomarkers and prognosis was explored, and a geriatric ESCC-related oxidative stress score (OSS) was constructed. To construct a predictive model for 3-year overall survival (OS), machine-learning strategies including decision tree (DT), random forest (RF), and support vector machine (SVM) were employed. These machine-learning strategies play a key role in data mining and pattern recognition tasks. Each model was tested in the external validation cohort through 1000 resampling iterations. Validation was conducted using receiver operating characteristic area under the curve (AUC) and calibration plots. RESULTS The training cohort and validation cohort consisted of 340 and 145 patients, respectively. In the training cohort, the 3-year OS rate for patients was 59.2%. We constructed the OSS based on systemic oxidative stress biomarkers using the training cohort. The study found that pathological N stage, pathological T stage, tumor histological type, lymphovascular invasion, CEA, OSS, CA 19 - 9, and the amount of bleeding were the most important factors influencing the 3-year OS. These eight important features were included in training the RF, DT, and SVM and trained on the training cohort and validated cohort, respectively. In the training cohort, the RF model demonstrated the highest predictive performance with an AUC of 0.975 (0.962-0.987), while the DT model is 0.784 (0.739-0.830) and the SVM is 0.879 (0.843-0.916). In the external validation cohort, the RF model again exhibited the highest performance with an AUC of 0.791 (0.717-0.864), compared to the DT model with an AUC of 0.717 (0.640-0.794) and 0.779 (0.702-0.856) in SVM. CONCLUSIONS The random forest clinical prediction model constructed based on OSS can effectively predict the prognosis of elderly patients with ESCC after curative surgery.
Collapse
Affiliation(s)
- Jin-Biao Xie
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Putian University, No.999 Dongzhen Road, Fujian, 351100, China.
| | - Shi-Jie Huang
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Putian University, No.999 Dongzhen Road, Fujian, 351100, China
| | - Tian-Bao Yang
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Putian University, No.999 Dongzhen Road, Fujian, 351100, China
| | - Wu Wang
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Putian University, No.999 Dongzhen Road, Fujian, 351100, China
| | - Bo-Yang Chen
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Putian University, No.999 Dongzhen Road, Fujian, 351100, China
| | - Lianyi Guo
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| |
Collapse
|
5
|
Hu J, Cai Y, Chen Y, Zhu X. Serum Direct Bilirubin as a Biomarker for Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:735-743. [PMID: 39530054 PMCID: PMC11552383 DOI: 10.2147/bctt.s491523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Background The role of serum total bilirubin (TB) in cancer has been a subject of controversy, as has the role of its subtypes, particularly serum direct bilirubin (DB). The aim of the present study was to investigate the association between serum DB levels and breast cancer, as well as to assess the diagnostic utility of serum DB in breast cancer. Methods A total of 5299 patients diagnosed with breast cancer for the first time at Taizhou Hospital of Zhejiang Province were included in the study, and 10028 healthy physical examination subjects were included as healthy controls. Logistics regression was used to investigate the relationship between serum DB and breast cancer, and the value of serum DB in the diagnosis of breast cancer was assessed by means of receiver operator characteristic (ROC) curve analysis. Results The serum DB concentration in the breast cancer group was significantly higher than the healthy controls (P < 0.001). Multivariate logistic regression results show that serum DB was an independent risk factor for breast cancer (odds ratio [OR]=4.504, 95% confidence interval [CI]: 4.200-4.831). Subjects with a serum DB concentration in the fourth quartile had a higher risk of breast cancer occurrence compared to those in the first quartile after adjusting for age (OR = 7.155, 95%CI: 6.474-7.907). The optimal cut-off value of serum DB for diagnosing breast cancer was determined to be 2.75 μmol/L, with an area under the curve (AUC) of 0.712 (95% CI: 0.703-0.722). This value exhibited good specificity (77.0%) and negative predictive value (77.8%). Conclusion Serum DB was identified as a risk factor for breast cancer, demonstrating good diagnostic potential for the disease. These findings suggest that serum DB could serve as a promising serum molecular marker for breast cancer.
Collapse
Affiliation(s)
- Jinxi Hu
- Department of Oncological Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, People’s Republic of China
| | - Yangjun Cai
- Department of Oncological Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, People’s Republic of China
| | - Yijun Chen
- Department of Laboratory Medicine, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, People’s Republic of China
| | - Xiaoli Zhu
- Department of Laboratory Medicine, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, People’s Republic of China
| |
Collapse
|
6
|
AlAseeri AA, Al-Kuraishy HM, Al-Gareeb AI, Ali NH, Alexiou A, Papadakis M, Bahaa MM, Alruwaili M, Batiha GES. The compelling role of allopurinol in hyperuricemia-induced epilepsy: Unrecognized like tears in rain. Brain Res Bull 2024; 213:110973. [PMID: 38723694 DOI: 10.1016/j.brainresbull.2024.110973] [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: 01/28/2024] [Revised: 04/28/2024] [Accepted: 05/02/2024] [Indexed: 05/23/2024]
Abstract
Epilepsy is a common neurological disease characterized by the recurrent, paroxysmal, and unprovoked seizures. It has been shown that hyperuricemia enhances and associated with the development and progression of epilepsy through induction of inflammation and oxidative stress. In addition, uric acid is released within the brain and contributes in the development of neuronal hyperexcitability and epileptic seizure. Brain uric acid acts as damage associated molecular pattern (DAMP) activates the immune response and induce the development of neuroinflammation. Therefore, inhibition of xanthine oxidase by allopurinol may reduce hyperuricemia-induced epileptic seizure and associated oxidative stress and inflammation. However, the underlying mechanism of allopurinol in the epilepsy was not fully elucidated. Therefore, this review aims to revise from published articles the link between hyperuricemia and epilepsy, and how allopurinol inhibits the development of epileptic seizure.
Collapse
Affiliation(s)
- Ali Abdullah AlAseeri
- Department of Internal Medicine, College of Medicine, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Naif H Ali
- Department of Internal Medicine, Medical College, Najran University, Najran, Saudi Arabia
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India; Department of Research & Development, Funogen, Athens 11741, Greece; Department of Research & Development, AFNP Med, Wien 1030, Austria; Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW 2770, Australia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, Wuppertal 42283, Germany.
| | - Mostafa M Bahaa
- Pharmacy Practice Department, Faculty of Pharmacy, Horus University, New Damietta, Egypt
| | - Mubarak Alruwaili
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira 22511, Egypt
| |
Collapse
|
7
|
Huang Z, Zhou Z, Ma Y, Hu YM. Mito-Tempo alleviates ox-LDL-provoked foam cell formation by regulating Nrf2/NLRP3 signaling. Biosci Biotechnol Biochem 2024; 88:759-767. [PMID: 38719485 DOI: 10.1093/bbb/zbae058] [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: 11/30/2023] [Accepted: 04/25/2024] [Indexed: 06/22/2024]
Abstract
Our previous studies have demonstrated that Mito-Tempol (also known as 4-hydroxy-Tempo), a mitochondrial reactive oxygen species scavenger, alleviates oxidized low-density lipoprotein (ox-LDL)-triggered foam cell formation. Given the effect of oxidative stress on activating the NOD-, LRR-, and pyrin domain-containing 3 (NLRP3) inflammasome, which promotes foam cell formation, we aimed to explore whether Mito-Tempo inhibits ox-LDL-triggered foam cell formation by regulating NLRP3 inflammasome. The results revealed that Mito-Tempo re-activated Nrf2 and alleviated macrophage foam cell formation induced by ox-LDL, whereas the effects were reversed by ML385 (a specific Nrf2 inhibitor). Mito-Tempo restored the expression and nuclear translocation of Nrf2 by decreasing ox-LDL-induced ubiquitination. Furthermore, Mito-Tempo suppressed ox-LDL-triggered NLRP3 inflammasome activation and subsequent pyroptosis, whereas the changes were blocked by ML385. Mito-Tempo decreased lipoprotein uptake by inhibiting CD36 expression and suppressed foam cell formation by regulating the NLRP3 inflammasome. Taken together, Mito-Tempo exhibits potent anti-atherosclerotic effects by regulating Nrf2/NLRP3 signaling.
Collapse
Affiliation(s)
- Zhenyu Huang
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhaoli Zhou
- Shanghai Key Laboratory for Molecular Imaging, Collaborative Scientific Research Center, Shanghai University of Medicine & Health Science, Shanghai, China
- Department of Pharmacology, School of Pharmacy, Shanghai University of Medicine & Health Science, Shanghai, China
| | - Ying Ma
- Department of Geriatrics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yao-Min Hu
- Department of Geriatrics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
8
|
Li S, Shi Y, Zhu J, Li J, Wang S, Liu C. Protective effect of oxytocin on vincristine-induced gastrointestinal dysmotility in mice. Front Pharmacol 2024; 15:1270612. [PMID: 38655179 PMCID: PMC11037254 DOI: 10.3389/fphar.2024.1270612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 02/26/2024] [Indexed: 04/26/2024] Open
Abstract
Aims: Vincristine (VCR), an antineoplastic drug, induces peripheral neuropathy characterized by nerve damage, limiting its use and reducing the quality of life of patients. VCR causes myenteric neuron damage, inhibits gastrointestinal motility, and results in constipation or paralytic ileus in patients. Oxytocin (OT) is an endogenous neuropeptide produced by the enteric nerve system, which regulates gastrointestinal motility and exerts neuroprotective effects. This study aimed to investigate whether OT can improve VCR-induced gastrointestinal dysmotility and evaluate the underlying mechanism. Methods: Mice were injected either with saline or VCR (0.1 mg/kg/d, i. p.) for 14 days, and OT (0.1 mg/kg/d, i.p.) was applied 1 h before each VCR injection. Gastrointestinal transit and the contractile activity of the isolated colonic segments were assessed. The concentration of OT in plasma was measured using ELISA. Immunofluorescence staining was performed to analyze myenteric neurons and reactive oxygen species (ROS) levels. Furthermore, the indicators of oxidative stress were detected. The protein expressions of Nrf2, ERK1/2, P-ERK1/2, p38, and P-p38 in the colon were tested using Western blot. Results: VCR reduced gastrointestinal transit and the responses of isolated colonic segments to electrical field stimulation and decreased the amount of neurons. Furthermore, VCR reduced neuronal nitric oxide synthase and choline acetyltransferase immunopositive neurons in the colonic myenteric nerve plexus. VCR increased the concentration of OT in plasma. Exogenous OT pretreatment ameliorated the inhibition of gastrointestinal motility and the injury of myenteric neurons caused by VCR. OT pretreatment also prevented the decrease of superoxide dismutase activity, glutathione content, total antioxidative capacity, and Nrf2 expression, the increase of ROS levels, and the phosphorylation of ERK1/2 and p38 MAPK following VCR treatment. Conclusion: Our results suggest that OT pretreatment can protect enteric neurons from VCR-induced injury by inhibiting oxidative stress and MAPK pathways (ERK1/2, p38). This may be the underlying mechanism by which it alleviates gastrointestinal dysmotility.
Collapse
Affiliation(s)
- Shuang Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, China
| | - Yao Shi
- Ministry of Education Key Laboratory of Protein, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jianchun Zhu
- Department of Physiology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, China
| | - Jingxin Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, China
| | - Shuanglian Wang
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Chuanyong Liu
- Department of Physiology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, China
- Provincial Key Lab of Mental Disorders, Shandong University, Jinan, China
| |
Collapse
|
9
|
Wang X, Yan J, Ni X, Hu S, Zhang M, Ying Y. Phloretin targets SIRT1 to alleviate oxidative stress, apoptosis, and inflammation in deep venous thrombosis. Toxicol Res 2024; 40:83-96. [PMID: 38223667 PMCID: PMC10786814 DOI: 10.1007/s43188-023-00207-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/04/2023] [Accepted: 08/14/2023] [Indexed: 01/16/2024] Open
Abstract
Deep vein thrombosis (DVT) is a type of venous thromboembolism posing a serious threat to health on a global scale. Phloretin is a potential natural product that has a variety of pharmacological activities. Besides, some Chinese medicines reported that deacetylase sirtuin (SIRT)1 treats DVT by anti-inflammatory and anti-platelet production. However, the specific binding targets and binding modes have not been elaborated. The present study was to investigate whether phloretin attenuates DVT in model rats and oxidized low‑density lipoprotein (ox‑LDL) induced human umbilical vein endothelial cells (HUVECs), and to explore its potential target. The results revealed that the treatment of phloretin, especially pretreatment of it elevated tissue plasminogen activator (t-PA), superoxide dismutase (SOD), prothrombin time (PT), thrombin time (TT), activated partial thromboplastin time (APTT), and cell apoptosis proteins whereas it suppressed plasminogen activator inhibitor (PAI), malondialdehyde (MDA), reactive oxygen species (ROS), fibrinogen (FIB) in DVT rats and cells. Concurrently, phloretin inhibited collagen type I alpha 1 (COL1A1), transforming growth factor-β1 (TGF-β1), and inflammatory factors while it enhanced nuclear factor erythroid 2-related factor 2 (Nrf-2), heme oxygenase 1 (HO-1). In addition, 20 μM phloretin exerted powerful effective protection in HUVECs with DVT model. Later, the surface plasmon resonance (SPR) confirmed that phloretin has a high affinity with SIRT1. Furthermore, siRNA-SIRT1 transfection abolished the protective effect of phloretin against ox‑LDL‑induced DVT in HUVECs, indicating that phloretin targets SIRT1 to alleviate oxidative stress, cell apoptosis, and inflammation in DVT rats and HUVECs. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-023-00207-y.
Collapse
Affiliation(s)
- Xiaodong Wang
- Department of Vascular Surgery, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang China
- Department of Vascular Surgery, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang China
| | - Jin Yan
- Department of Vascular Surgery, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang China
- Department of Vascular Surgery, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang China
| | - Xiaolong Ni
- Department of Vascular Surgery, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang China
- Department of Vascular Surgery, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang China
| | - Sipin Hu
- Department of Vascular Surgery, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang China
- Department of Vascular Surgery, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang China
| | - Mingwan Zhang
- Department of Pharmacy, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang China
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang China
| | - Yin Ying
- Department of Pharmacy, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang China
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang China
| |
Collapse
|
10
|
Ng D, Pawling J, Dennis JW. Gene purging and the evolution of Neoave metabolism and longevity. J Biol Chem 2023; 299:105409. [PMID: 37918802 PMCID: PMC10722388 DOI: 10.1016/j.jbc.2023.105409] [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: 09/11/2023] [Revised: 10/19/2023] [Accepted: 10/25/2023] [Indexed: 11/04/2023] Open
Abstract
Maintenance of the proteasome requires oxidative phosphorylation (ATP) and mitigation of oxidative damage, in an increasingly dysfunctional relationship with aging. SLC3A2 plays a role on both sides of this dichotomy as an adaptor to SLC7A5, a transporter of branched-chain amino acids (BCAA: Leu, Ile, Val), and to SLC7A11, a cystine importer supplying cysteine to the synthesis of the antioxidant glutathione. Endurance in mammalian muscle depends in part on oxidation of BCAA; however, elevated serum levels are associated with insulin resistance and shortened lifespans. Intriguingly, the evolution of modern birds (Neoaves) has entailed the purging of genes including SLC3A2, SLC7A5, -7, -8, -10, and SLC1A4, -5, largely removing BCAA exchangers and their interacting Na+/Gln symporters in pursuit of improved energetics. Additional gene purging included mitochondrial BCAA aminotransferase (BCAT2), pointing to reduced oxidation of BCAA and increased hepatic conversion to triglycerides and glucose. Fat deposits are anhydrous and highly reduced, maximizing the fuel/weight ratio for prolonged flight, but fat accumulation in muscle cells of aging humans contributes to inflammation and senescence. Duplications of the bidirectional α-ketoacid transporters SLC16A3, SLC16A7, the cystine transporters SLC7A9, SLC7A11, and N-glycan branching enzymes MGAT4B, MGAT4C in Neoaves suggests a shift to the transport of deaminated essential amino acid, and stronger mitigation of oxidative stress supported by the galectin lattice. We suggest that Alfred Lotka's theory of natural selection as a maximum power organizer (PNAS 8:151,1922) made an unusually large contribution to Neoave evolution. Further molecular analysis of Neoaves may reveal novel rewiring with applications for human health and longevity.
Collapse
Affiliation(s)
- Deanna Ng
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Judy Pawling
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - James W Dennis
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto Ontario, Canada.
| |
Collapse
|
11
|
Ma P, Zhao M, Li Y, Zhang G, Ma Y, Shi Y, Su P, Chen R, Tang ZG, Zhang Y, Liu B, Zhang Q, Liu X, Li F. The protective effects of uric acid against myocardial ischemia via the Nrf2 pathway. Eur J Pharmacol 2023; 959:176062. [PMID: 37793494 DOI: 10.1016/j.ejphar.2023.176062] [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: 06/21/2023] [Revised: 09/03/2023] [Accepted: 09/18/2023] [Indexed: 10/06/2023]
Abstract
Uric acid (UA) possesses both pro- and anti-oxidative properties in ischemic heart disease, but the underlying mechanism remains unclear. We aimed to investigate UA's protective effect on myocardial ischemia by examining its effects on ECG Ischemic Alterations (EIA) and H2O2-induced oxidative stress in H9C2 myocardial cells. The incidence of EIA decreased over time and was more prevalent among women than men. A U-shaped relationship was observed between UA levels and EIA incidence, with the third quartile exhibiting a protective association. Addition of 237.9 μmol/L UA improved cellular damage and oxidative stress in H2O2-treated H9C2 cells, as determined by cell viability, LDH release, ROS levels, and total antioxidant capacity assays. UA activated the Nrf2 pathway, evidenced by increased expression of Nrf2, GCLC, and HO-1 proteins. By reversing cell cycle blockage, promoting wound healing ability, improving colony-forming capacity, and increasing angiogenesis in H2O2-treated cells, UA exhibited positive effects on cardiomyocyte growth characteristics. Additionally, use of Nrf2 inhibitor ML385 confirmed the involvement of the Nrf2 pathway by negating UA's effects on oxidatively damaged cardiomyocytes. Our findings suggest that UA induces downstream antioxidant factors to ameliorate oxidative stress by activating the Nrf2 pathway, which could be one of the targets responsible for UA's beneficial effects in myocardial ischemia.
Collapse
Affiliation(s)
- Pengqiang Ma
- Department of Preventive Medicine, School of Public Health, Hubei University of Medicine, Shiyan, China; Hubei Biomedical Detection Sharing Platform in Water Source Area of South to North Water Diversion Project, Hubei University of Medicine, Shiyan, China
| | - Menghui Zhao
- Department of Preventive Medicine, School of Public Health, Hubei University of Medicine, Shiyan, China; Hubei Biomedical Detection Sharing Platform in Water Source Area of South to North Water Diversion Project, Hubei University of Medicine, Shiyan, China
| | - Yue Li
- Department of Preventive Medicine, School of Public Health, Hubei University of Medicine, Shiyan, China; Hubei Biomedical Detection Sharing Platform in Water Source Area of South to North Water Diversion Project, Hubei University of Medicine, Shiyan, China
| | - Guanqian Zhang
- Department of Preventive Medicine, School of Public Health, Hubei University of Medicine, Shiyan, China; Hubei Biomedical Detection Sharing Platform in Water Source Area of South to North Water Diversion Project, Hubei University of Medicine, Shiyan, China
| | - Yuxia Ma
- Department of Preventive Medicine, School of Public Health, Hubei University of Medicine, Shiyan, China; Hubei Biomedical Detection Sharing Platform in Water Source Area of South to North Water Diversion Project, Hubei University of Medicine, Shiyan, China
| | - Youlan Shi
- Department of Preventive Medicine, School of Public Health, Hubei University of Medicine, Shiyan, China; Hubei Biomedical Detection Sharing Platform in Water Source Area of South to North Water Diversion Project, Hubei University of Medicine, Shiyan, China
| | - Peihui Su
- Department of Preventive Medicine, School of Public Health, Hubei University of Medicine, Shiyan, China; Hubei Biomedical Detection Sharing Platform in Water Source Area of South to North Water Diversion Project, Hubei University of Medicine, Shiyan, China
| | - Rongxia Chen
- Department of Preventive Medicine, School of Public Health, Hubei University of Medicine, Shiyan, China; Hubei Biomedical Detection Sharing Platform in Water Source Area of South to North Water Diversion Project, Hubei University of Medicine, Shiyan, China; Department of Drug Quality Inspection, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, China
| | - Zhen-Gang Tang
- Health Management Center, Shiyan Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Yan Zhang
- Health Management Center, Shiyan Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Bing Liu
- Department of Preventive Medicine, School of Public Health, Hubei University of Medicine, Shiyan, China
| | - Qiong Zhang
- Department of Preventive Medicine, School of Public Health, Hubei University of Medicine, Shiyan, China; Hubei Biomedical Detection Sharing Platform in Water Source Area of South to North Water Diversion Project, Hubei University of Medicine, Shiyan, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China; Department of Drug Quality Inspection, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, China.
| | - Xiang Liu
- Health Management Center, Shiyan Renmin Hospital, Hubei University of Medicine, Shiyan, China.
| | - Feifeng Li
- Department of Preventive Medicine, School of Public Health, Hubei University of Medicine, Shiyan, China; Hubei Biomedical Detection Sharing Platform in Water Source Area of South to North Water Diversion Project, Hubei University of Medicine, Shiyan, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China; Department of Drug Quality Inspection, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, China; Health Management Center, Shiyan Renmin Hospital, Hubei University of Medicine, Shiyan, China.
| |
Collapse
|
12
|
Fan K, Sun T, Yin F. J-shaped association between uric acid and breast cancer risk: a prospective case-control study. J Cancer Res Clin Oncol 2023; 149:7629-7636. [PMID: 36995406 PMCID: PMC10374747 DOI: 10.1007/s00432-023-04725-y] [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: 03/07/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023]
Abstract
BACKGROUND/AIM In terms of breast cancer risk, there is no consensus on the effect of uric acid (UA) levels. The aim of our study was to clarify the link between UA and breast cancer risk in a prospective case-control study and to find the UA threshold point. METHODS We designed a case-control study with 1050 females (525 newly diagnosed breast cancer patients and 525 controls). We measured the UA levels at baseline and confirmed the incidence of breast cancer through postoperative pathology. We used binary logistic regression to study the association between breast cancer and UA. In addition, we performed restricted cubic splines to evaluate the potential nonlinear links between UA and breast cancer risk. We used threshold effect analysis to identify the UA cut-off point. RESULTS After adjusting for multiple confounding factors, we found that compared with the referential level (3.5-4.4 mg/dl), the odds ratio (OR) of breast cancer was 1.946 (95% CI 1.140-3.321) (P < 0.05) in the lowest UA level and 2.245 (95% CI 0.946-5.326) (P > 0.05) in the highest level. Using the restricted cubic bar diagram, we disclosed a J-shaped association between UA and breast cancer risk (P-nonlinear < 0.05) after adjusting for all confounders. In our study, 3.6 mg/dl was found to be the UA threshold which acted as the optimal turning point of the curve. The OR for breast cancer was 0.170 (95% CI 0.056-0.512) to the left and 1.283 (95% CI 1.074-1.532) to the right of 3.6 mg/dl UA (P for log likelihood ratio test < 0.05). CONCLUSION We found a J-shaped association between UA and breast cancer risk. Controlling the UA level around the threshold point of 3.6 mg/dl provides a novel insight into breast cancer prevention.
Collapse
Affiliation(s)
- Kexin Fan
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Pneumology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei, China
| | - Tengfei Sun
- Department of Gastrology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei, China
| | - Fuzai Yin
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China.
- Department of Endocrinology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei, China.
| |
Collapse
|
13
|
Čypienė A, Gimžauskaitė S, Rinkūnienė E, Jasiūnas E, Laucevičius A, Ryliškytė L, Badarienė J. Effect of Alcohol Consumption Habits on Early Arterial Aging in Subjects with Metabolic Syndrome and Elevated Serum Uric Acid. Nutrients 2023; 15:3346. [PMID: 37571284 PMCID: PMC10421141 DOI: 10.3390/nu15153346] [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: 07/04/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Hyperuricemia is perceived as one of the risk factors for developing and progressing cardiovascular disease and metabolic syndrome through various pathological mechanisms. Endogenous synthesis and exogenous factors such as diet and beverages consumed play a major role in determining serum uric acid (sUA) levels. The aim of this study was to evaluate the effect of alcohol consumption on early arterial aging in middle-aged patients with metabolic syndrome (MetS) and hyperuricemia. MATERIALS AND METHODS This study included 661 middle-aged subjects (241 men and 420 women) from the Lithuanian High Cardiovascular Risk (LitHiR) primary prevention program. Characteristics of subjects such as blood pressure, laboratory testing, and the specialized nutrition profile questionnaire were evaluated. As an early marker of arterial stiffness, carotid-femoral pulse wave velocity (cfPWV) was assessed using a non-invasive applanation tonometry technique. RESULTS Hyperuricemia was present in 29% of men and 34% of women. Hyperuricemic men reported 1.6 times higher rates of alcohol drinking compared to men with normal sUA levels. After analyzing the correlation between alcohol consumption and cfPWV, no statistically significant relationships were found at a significance level of α = 0.05 but lowering the significance level to 0.06 revealed significant associations in men with normal sUA (ε2ordinal = 0.05, p = 0.06) and in women with increased sUA levels (ε2ordinal = 0.05, p = 0.08). Regression analysis showed that hyperuricemic men, consuming more than one unit of alcohol per week, had a significant impact on increasing cfPWV, while men with normal sUA levels, abstaining from alcohol entirely, resulted in a statistically significant decrease in cfPWV. Our results showed statistically significant relationships only among a group of men, although the women in the hyperuricemic group had a statistically higher cfPWV than women with normal sUA levels. CONCLUSIONS Drinking alcohol is associated with increased arterial stiffness among hyperuricemic middle-aged men with MetS.
Collapse
Affiliation(s)
- Alma Čypienė
- State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania; (A.Č.); (A.L.)
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (E.R.); (L.R.); (J.B.)
| | - Silvija Gimžauskaitė
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (E.R.); (L.R.); (J.B.)
| | - Egidija Rinkūnienė
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (E.R.); (L.R.); (J.B.)
| | - Eugenijus Jasiūnas
- Center of Informatics and Development, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania;
| | - Aleksandras Laucevičius
- State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania; (A.Č.); (A.L.)
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (E.R.); (L.R.); (J.B.)
| | - Ligita Ryliškytė
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (E.R.); (L.R.); (J.B.)
| | - Jolita Badarienė
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (E.R.); (L.R.); (J.B.)
| |
Collapse
|
14
|
Li K, Li K, Yao Q, Shui X, Zheng J, He Y, Lei W. The potential relationship of coronary artery disease and hyperuricemia: A cardiometabolic risk factor. Heliyon 2023; 9:e16097. [PMID: 37215840 PMCID: PMC10199191 DOI: 10.1016/j.heliyon.2023.e16097] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 05/24/2023] Open
Abstract
Coronary arterial disease (CAD) is the leading cause of mortality in the world. Hyperuricemia has recently emerged as a novel independent risk factor of CAD, in addition to the traditional risk factors such as hyperlipidemia, smoking, and obesity. Several clinical studies have shown that hyperuricemia is strongly associated with the risk, progression and poor prognosis of CAD, as well as verifying an association with traditional CAD risk factors. Uric acid or enzymes in the uric acid production pathway are associated with inflammation, oxidative stress, regulation of multiple signaling pathways and the renin-angiotensin-aldosterone system (RAAS), and these pathophysiological alterations are currently the main mechanisms of coronary atherosclerosis formation. The risk of death from CAD can be effectively reduced by the uric acid-lowering therapy, but the interventional treatment of uric acid levels in patients with CAD remains controversial due to the diversity of co-morbidities and the complexity of causative factors. In this review, we analyze the association between hyperuricemia and CAD, elucidate the possible mechanisms by which uric acid induces or exacerbates CAD, and discuss the benefits and drawbacks of uric acid-lowering therapy. This review could provide theoretical references for the prevention and management of hyperuricemia-associated CAD.
Collapse
Affiliation(s)
- Kaiyue Li
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Department of Precision Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Kongwei Li
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Department of Precision Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
- Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Qingmei Yao
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Department of Precision Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Xiaorong Shui
- Laboratory of Vascular Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Jing Zheng
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI, USA
| | - Yuan He
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Department of Precision Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Wei Lei
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Department of Precision Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
15
|
de Lima JD, de Paula AGP, Yuasa BS, de Souza Smanioto CC, da Cruz Silva MC, Dos Santos PI, Prado KB, Winter Boldt AB, Braga TT. Genetic and Epigenetic Regulation of the Innate Immune Response to Gout. Immunol Invest 2023; 52:364-397. [PMID: 36745138 DOI: 10.1080/08820139.2023.2168554] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gout is a disease caused by uric acid (UA) accumulation in the joints, causing inflammation. Two UA forms - monosodium urate (MSU) and soluble uric acid (sUA) have been shown to interact physically with inflammasomes, especially with the nod-like receptor (NLR) family pyrin domain containing 3 (NLRP3), albeit the role of the immune response to UA is poorly understood, given that asymptomatic hyperuricemia does also exist. Macrophage phagocytosis of UA activate NLRP3, lead to cytokines release, and ultimately, lead to chemoattract neutrophils and lymphocytes to the gout flare joint spot. Genetic variants of inflammasome genes and of genes encoding their molecular partners may influence hyperuricemia and gout susceptibility, while also influencing other comorbidities such as metabolic syndrome and cardiovascular diseases. In this review, we summarize the inflammatory responses in acute and chronic gout, specifically focusing on innate immune cell mechanisms and genetic and epigenetic characteristics of participating molecules. Unprecedently, a novel UA binding protein - the neuronal apoptosis inhibitor protein (NAIP) - is suggested as responsible for the asymptomatic hyperuricemia paradox.Abbreviation: β2-integrins: leukocyte-specific adhesion molecules; ABCG2: ATP-binding cassete family/breast cancer-resistant protein; ACR: American college of rheumatology; AIM2: absent in melanoma 2, type of pattern recognition receptor; ALPK1: alpha-protein kinase 1; ANGPTL2: angiopoietin-like protein 2; ASC: apoptosis-associated speck-like protein; BIR: baculovirus inhibitor of apoptosis protein repeat; BIRC1: baculovirus IAP repeat-containing protein 1; BIRC2: baculoviral IAP repeat-containing protein 2; C5a: complement anaphylatoxin; cAMP: cyclic adenosine monophosphate; CARD: caspase activation and recruitment domains; CARD8: caspase recruitment domain-containing protein 8; CASP1: caspase 1; CCL3: chemokine (C-C motif) ligand 3; CD14: cluster of differentiation 14; CD44: cluster of differentiation 44; Cg05102552: DNA-methylation site, usually cytosine followed by guanine nucleotides; contains arbitrary identification code; CIDEC: cell death-inducing DNA fragmentation factor-like effector family; CKD: chronic kidney disease; CNV: copy number variation; CPT1A: carnitine palmitoyl transferase - type 1a; CXCL1: chemokine (CXC motif) ligand 1; DAMPs: damage associated molecular patterns; DC: dendritic cells; DNMT(1): maintenance DNA methyltransferase; eQTL: expression quantitative trait loci; ERK1: extracellular signal-regulated kinase 1; ERK2: extracellular signal-regulated kinase 2; EULAR: European league against rheumatism; GMCSF: granulocyte-macrophage colony-stimulating factor; GWAS: global wide association studies; H3K27me3: tri-methylation at the 27th lysine residue of the histone h3 protein; H3K4me1: mono-methylation at the 4th lysine residue of the histone h3 protein; H3K4me3: tri-methylation at the 4th lysine residue of the histone h3 protein; HOTAIR: human gene located between hoxc11 and hoxc12 on chromosome 12; IκBα: cytoplasmatic protein/Nf-κb transcription inhibitor; IAP: inhibitory apoptosis protein; IFNγ: interferon gamma; IL-1β: interleukin 1 beta; IL-12: interleukin 12; IL-17: interleukin 17; IL18: interleukin 18; IL1R1: interleukin-1 receptor; IL-1Ra: interleukin-1 receptor antagonist; IL-22: interleukin 22; IL-23: interleukin 23; IL23R: interleukin 23 receptor; IL-33: interleukin 33; IL-6: interleukin 6; IMP: inosine monophosphate; INSIG1: insulin-induced gene 1; JNK1: c-jun n-terminal kinase 1; lncRNA: long non-coding ribonucleic acid; LRR: leucine-rich repeats; miR: mature non-coding microRNAs measuring from 20 to 24 nucleotides, animal origin; miR-1: miR followed by arbitrary identification code; miR-145: miR followed by arbitrary identification code; miR-146a: miR followed by arbitrary identification code, "a" stands for mir family; "a" family presents similar mir sequence to "b" family, but different precursors; miR-20b: miR followed by arbitrary identification code; "b" stands for mir family; "b" family presents similar mir sequence to "a" family, but different precursors; miR-221: miR - followed by arbitrary identification code; miR-221-5p: miR followed by arbitrary identification code; "5p" indicates different mature miRNAs generated from the 5' arm of the pre-miRNA hairpin; miR-223: miR followed by arbitrary identification code; miR-223-3p: mir followed by arbitrary identification code; "3p" indicates different mature miRNAs generated from the 3' arm of the pre-miRNA hairpin; miR-22-3p: miR followed by arbitrary identification code, "3p" indicates different mature miRNAs generated from the 3' arm of the pre-miRNA hairpin; MLKL: mixed lineage kinase domain-like pseudo kinase; MM2P: inductor of m2-macrophage polarization; MSU: monosodium urate; mTOR: mammalian target of rapamycin; MyD88: myeloid differentiation primary response 88; n-3-PUFAs: n-3-polyunsaturated fatty-acids; NACHT: acronym for NAIP (neuronal apoptosis inhibitor protein), C2TA (MHC class 2 transcription activator), HET-E (incompatibility locus protein from podospora anserina) and TP1 (telomerase-associated protein); NAIP: neuronal apoptosis inhibitory protein (human); Naip1: neuronal apoptosis inhibitory protein type 1 (murine); Naip5: neuronal apoptosis inhibitory protein type 5 (murine); Naip6: neuronal apoptosis inhibitory protein type 6 (murine); NBD: nucleotide-binding domain; Nek7: smallest NIMA-related kinase; NET: neutrophil extracellular traps; Nf-κB: nuclear factor kappa-light-chain-enhancer of activated b cells; NFIL3: nuclear-factor, interleukin 3 regulated protein; NIIMA: network of immunity in infection, malignancy, and autoimmunity; NLR: nod-like receptor; NLRA: nod-like receptor NLRA containing acidic domain; NLRB: nod-like receptor NLRA containing BIR domain; NLRC: nod-like receptor NLRA containing CARD domain; NLRC4: nod-like receptor family CARD domain containing 4; NLRP: nod-like receptor NLRA containing PYD domain; NLRP1: nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing 1; NLRP12: nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing 12; NLRP3: nod-like receptor family pyrin domain containing 3; NOD2: nucleotide-binding oligomerization domain; NRBP1: nuclear receptor-binding protein; Nrf2: nuclear factor erythroid 2-related factor 2; OR: odds ratio; P2X: group of membrane ion channels activated by the binding of extracellular; P2X7: p2x purinoceptor 7 gene; p38: member of the mitogen-activated protein kinase family; PAMPs: pathogen associated molecular patters; PBMC: peripheral blood mononuclear cells; PGGT1B: geranylgeranyl transferase type-1 subunit beta; PHGDH: phosphoglycerate dehydrogenase; PI3-K: phospho-inositol; PPARγ: peroxisome proliferator-activated receptor gamma; PPARGC1B: peroxisome proliferative activated receptor, gamma, coactivator 1 beta; PR3: proteinase 3 antigen; Pro-CASP1: inactive precursor of caspase 1; Pro-IL1β: inactive precursor of interleukin 1 beta; PRR: pattern recognition receptors; PYD: pyrin domain; RAPTOR: regulatory associated protein of mTOR complex 1; RAS: renin-angiotensin system; REDD1: regulated in DNA damage and development 1; ROS: reactive oxygen species; rs000*G: single nuclear polymorphism, "*G" is related to snp where replaced nucleotide is guanine, usually preceded by an id number; SLC2A9: solute carrier family 2, member 9; SLC7A11: solute carrier family 7, member 11; SMA: smooth muscular atrophy; Smac: second mitochondrial-derived activator of caspases; SNP: single nuclear polymorphism; Sp3: specificity protein 3; ST2: serum stimulation-2; STK11: serine/threonine kinase 11; sUA: soluble uric acid; Syk: spleen tyrosine kinase; TAK1: transforming growth factor beta activated kinase; Th1: type 1 helper T cells; Th17: type 17 helper T cells; Th2: type 2 helper T cells; Th22: type 22 helper T cells; TLR: tool-like receptor; TLR2: toll-like receptor 2; TLR4: toll-like receptor 4; TNFα: tumor necrosis factor alpha; TNFR1: tumor necrosis factor receptor 1; TNFR2: tumor necrosis factor receptor 2; UA: uric acid; UBAP1: ubiquitin associated protein; ULT: urate-lowering therapy; URAT1: urate transporter 1; VDAC1: voltage-dependent anion-selective channel 1.
Collapse
Affiliation(s)
- Jordana Dinorá de Lima
- Microbiology, Parasitology and Pathology Program, Universidade Federal do Parana (UFPR), Curitiba, Brazil
| | | | - Bruna Sadae Yuasa
- Microbiology, Parasitology and Pathology Program, Universidade Federal do Parana (UFPR), Curitiba, Brazil
| | | | - Maria Clara da Cruz Silva
- Microbiology, Parasitology and Pathology Program, Universidade Federal do Parana (UFPR), Curitiba, Brazil
| | | | - Karin Braun Prado
- Genetics Program, Universidade Federal do Parana (UFPR), Curitiba, Brazil
| | - Angelica Beate Winter Boldt
- Program of Internal Medicine, Universidade Federal do Parana (UFPR), Curitiba, Brazil
- Genetics Program, Universidade Federal do Parana (UFPR), Curitiba, Brazil
| | - Tárcio Teodoro Braga
- Microbiology, Parasitology and Pathology Program, Universidade Federal do Parana (UFPR), Curitiba, Brazil
- Biosciences and Biotechnology Program, Instituto Carlos Chagas (ICC), Fiocruz-Parana, Brazil
| |
Collapse
|
16
|
Oxyresveratrol from mulberry branch extract protects HUVECs against oxidized Low-density Lipoprotein-induced oxidative injury via activation of the Nrf-2/HO-1 pathway. J Funct Foods 2023. [DOI: 10.1016/j.jff.2022.105371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
17
|
Wu Y, Liu J, Liu S, Fan W, Ding C, Gao Z, Tang Z, Luo Y, Shi X, Tan L, Song S. Bromoacetic acid causes oxidative stress and uric acid metabolism dysfunction via disturbing mitochondrial function and Nrf2 pathway in chicken kidney. ENVIRONMENTAL TOXICOLOGY 2022; 37:2910-2923. [PMID: 36017758 DOI: 10.1002/tox.23647] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/09/2022] [Accepted: 08/15/2022] [Indexed: 06/15/2023]
Abstract
Since the outbreak of COVID-19, widespread utilization of disinfectants has led to a tremendous increase in the generation of disinfection byproducts worldwide. Bromoacetic acid (BAA), one of the common disinfection byproducts in the environment, has triggered public concern because of its adverse effects on urinary system in mammals. Nevertheless, the BAA-induced nephrotoxicity and potential mechanism in birds still remains obscure. According to the detected content in the Taihu Lake Basin, the model of BAA exposure in chicken was established at doses of 0, 3, 300, 3000 μg/L for 4 weeks. Our results indicated that BAA exposure caused kidney swelling and structural disarrangement. BAA led to disorder in renal function (CRE, BUN, UA) and increased apoptosis (Bax, Bcl-2, caspase3). BAA suppressed the expression of mitochondrial biogenesis genes (PGC-1α, Nrf1, TFAM) and OXPHOS complex I genes (ND1, ND2, ND3, ND4, ND4L, ND5, ND6). Subsequently, BAA destroyed the expression of Nrf2 antioxidant reaction genes (Nrf2, Keap1, HO-1, NQO1, GCLM, GCLC). Furthermore, renal oxidative damage led to disorder in uric acid metabolism genes (Mrp2, Mrp4, Bcrp, OAT1, OAT2, OAT3) and exacerbated destruction in renal function. Overall, our study provided insights into the potential mechanism of BAA-induced nephrotoxicity, which were important for the clinical monitoring and prevention of BAA.
Collapse
Affiliation(s)
- Yuting Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jiwen Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Shuhui Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Wentao Fan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Chenchen Ding
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Zhangshan Gao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Zhihui Tang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yan Luo
- Administration for Market Regulation of Guangdong Province Key Laboratory of Supervision for Edible Agricultural Products, Shenzhen Centre of Inspection and Testing for Agricultural Products, Shenzhen, China
| | - Xizhi Shi
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo, China
| | - Lei Tan
- Administration for Market Regulation of Guangdong Province Key Laboratory of Supervision for Edible Agricultural Products, Shenzhen Centre of Inspection and Testing for Agricultural Products, Shenzhen, China
| | - Suquan Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
18
|
Hernández-Díazcouder A, González-Ramírez J, Sanchez F, Leija-Martínez JJ, Martínez-Coronilla G, Amezcua-Guerra LM, Sánchez-Muñoz F. Negative Effects of Chronic High Intake of Fructose on Lung Diseases. Nutrients 2022; 14:nu14194089. [PMID: 36235741 PMCID: PMC9571075 DOI: 10.3390/nu14194089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 11/09/2022] Open
Abstract
In the modern diet, excessive fructose intake (>50 g/day) had been driven by the increase, in recent decades, of the consumption of sugar-sweetened beverages. This phenomenon has dramatically increased within the Caribbean and Latin American regions. Epidemiological studies show that chronic high intake of fructose related to sugar-sweetened beverages increases the risk of developing several non-communicable diseases, such as chronic obstructive pulmonary disease and asthma, and may also contribute to the exacerbation of lung diseases, such as COVID-19. Evidence supports several mechanisms—such as dysregulation of the renin−angiotensin system, increased uric acid production, induction of aldose reductase activity, production of advanced glycation end-products, and activation of the mTORC1 pathway—that can be implicated in lung damage. This review addresses how these pathophysiologic and molecular mechanisms may explain the lung damage resulting from high intake of fructose.
Collapse
Affiliation(s)
| | - Javier González-Ramírez
- Cellular Biology Laboratory, Faculty of Nursing, Universidad Autónoma de Baja California Campus Mexicali, Mexicali 21100, Mexico
| | - Fausto Sanchez
- Department of Agricultural and Animal Production, Universidad Autónoma Metropolitana Xochimilco, Mexico City 04960, Mexico
| | - José J. Leija-Martínez
- Master and Doctorate Program in Medical, Dental, and Health Sciences, Faculty of Medicine, Universidad Nacional Autónoma de México Campus Ciudad Universitaria, Mexico City 04510, Mexico
- Research Laboratory of Pharmacology, Hospital Infantil de Mexico Federico Gómez, Mexico City 06720, Mexico
| | - Gustavo Martínez-Coronilla
- Histology Laboratory, Faculty of Medicine, Universidad Autónoma de Baja California Campus Mexicali, Mexicali 21100, Mexico
| | - Luis M. Amezcua-Guerra
- Department of Immunology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Fausto Sánchez-Muñoz
- Department of Immunology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
- Correspondence: ; Tel.: +52-5573-2911 (ext. 21310)
| |
Collapse
|
19
|
Patchouli Alcohol Inhibits D-Gal Induced Oxidative Stress and Ameliorates the Quality of Aging Cartilage via Activating the Nrf2/HO-1 Pathway in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6821170. [PMID: 35720186 PMCID: PMC9200550 DOI: 10.1155/2022/6821170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 03/07/2022] [Indexed: 12/15/2022]
Abstract
Chondrocytes play an essential role in maintaining the structure and function of articular cartilage. Oxidative stress occurred in chondrocytes accelerates cell senescence and death, which contributes to the development of osteoarthritis (OA). Patchouli alcohol (PA), a kind of sesquiterpene in Pogostemon cablin, processes multiple bioactivities in treatment of many diseases. However, its effects of antisenescence and antioxidation on chondrocytes in a D-gal-induced aging mice model are still obscure. In this study, we found that PA treatment could ameliorate the degradation of cartilage extracellular matrix (ECM) in a D-gal-induced aging mice model. Further analyses through the immunofluorescent staining and western blot revealed that PA inhibited D-gal-induced chondrocyte senescence via the activation of antioxidative system. Besides, the damage caused by D-gal could not be recovered with PA treatment in Nrf2-silencing chondrocytes. In addition, molecular docking analysis between PA and Keap1 further suggested that the mechanism of PA's antisenescence and antioxidation was attributed to the activation of Nrf2/HO-1 pathway. Therefore, our results demonstrated that PA was a promising candidate for preventing the quality loss of aging cartilage through inhibiting oxidative stress-mediated senescence in chondrocytes.
Collapse
|