1
|
Jiang Z, Zhu X, Zhao D, Jiang H, Wang X, Su F. Associations between non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio and hyperuricemia: a cross-sectional study. Lipids Health Dis 2024; 23:280. [PMID: 39227876 PMCID: PMC11370097 DOI: 10.1186/s12944-024-02269-9] [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: 06/18/2024] [Accepted: 08/20/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND AND OBJECTIVE The value of the non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio (NHHR) assessment in the context of metabolic abnormalities is growing in importance. Nevertheless, the relationship between NHHR and hyperuricemia (HUA) is unknown. This study seeks to investigate the relationship between NHHR and HUA. METHODS The data derived from the 2017-2020 National Health and Nutrition Examination Survey (NHANES) included 7,876 adult participants. The multivariable logistic regression model, subgroup analysis and smooth fitting curve were utilized in order to investigate the association between NHHR and HUA. RESULTS In the fully adjusted model 3, NHHR was significantly associated with HUA. Specifically, participants in the highest quartile of NHHR had 1.95 times higher odds of HUA prevalence compared to those in the lowest quartile [2.95 (2.39, 3.64), P < 0.0001]. Although the overall trend suggested a positive association, further analysis using smooth fitting curves and threshold effect analysis indicated that this association was nonlinear, with an inflection point at 5.8. The positive association persisted across different HUA definitions and after removing outliers. Subgroup analysis showed significant interactions between NHHR and HUA in different races and diabetes statuses. The odds of HUA prevalence were higher among non-diabetic participants [1.40 (1.32, 1.49), P < 0.0001] compared to diabetic participants [1.18 (1.06, 1.32), P = 0.0031]. Mexican Americans had the lowest odds of HUA prevalence [1.09 (0.92, 1.27), P = 0.2413] compared to other races. CONCLUSIONS There is a significant positive association between NHHR and HUA, indicating that NHHR may serve as a potential risk assessment maker for HUA, although further prospective studies are needed for validation.
Collapse
Affiliation(s)
- Zhimeng Jiang
- Graduate School of Hebei North University, Zhangjiakou, 075031, Hebei Province, China
- Department of Gastroenterology, Air Force Medical Center, Chinese People's Liberation Army, Beijing, Beijing, 100142, China
| | - Xingyu Zhu
- Graduate School of Hebei North University, Zhangjiakou, 075031, Hebei Province, China
- Department of Cardiovascular Medicine, Air Force Medical Center, Chinese People's Liberation Army, Beijing, Beijing, 100142, China
| | - Donglin Zhao
- Graduate School of Hebei North University, Zhangjiakou, 075031, Hebei Province, China
- Department of Gastroenterology, Air Force Medical Center, Chinese People's Liberation Army, Beijing, Beijing, 100142, China
| | - Huixin Jiang
- Graduate School of Kunming, Medical University Haiyuan College, Kunming, 65000, Yunnan Province, China
| | - Xiaoying Wang
- Department of Gastroenterology, Air Force Medical Center, Chinese People's Liberation Army, Beijing, Beijing, 100142, China
| | - Feifei Su
- Department of Cardiovascular Medicine, Air Force Medical Center, Chinese People's Liberation Army, Beijing, Beijing, 100142, China.
| |
Collapse
|
2
|
Sun T, Guo Y, Su Y, Shan S, Qian W, Zhang F, Li M, Zhang Z. Molecular mechanisms of diabetic nephropathy: A narrative review. Cell Biol Int 2024; 48:1240-1253. [PMID: 38946126 DOI: 10.1002/cbin.12212] [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: 04/23/2024] [Revised: 06/01/2024] [Accepted: 06/16/2024] [Indexed: 07/02/2024]
Abstract
Diabetic nephropathy (DN) is the predominant secondary nephropathy resulting in global end-stage renal disease. It is attracting significant attention in both domestic and international research due to its widespread occurrence, fast advancement, and limited choices for prevention and treatment. The pathophysiology of this condition is intricate and involves multiple molecular and cellular pathways at various levels. This article provides a concise overview of the molecular processes involved in the development of DN. It discusses various factors, such as signaling pathways, cytokines, inflammatory responses, oxidative stress, cellular damage, autophagy, and epigenetics. The aim is to offer clinicians a valuable reference for DN's diagnosis, treatment, and intervention.
Collapse
Affiliation(s)
- Tian Sun
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yina Guo
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yanting Su
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Shigang Shan
- School of Public Health and Nursing, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Wenbin Qian
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Feixue Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Mengxi Li
- School of Nuclear Technology and Chemistry & Biology, Hubei University of Science and Technology, Xianning, China
| | - Zhenwang Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
3
|
Uuh Narvaez JJ, Acevedo Fernández JJ, Negrete León E, Martínez Rivera JI, Guillermo Cordero JL, Segura Campos MR. Therapeutic Potential of Brassica oleracea and Raphanus sativus Aqueous Extracts on a Prediabetic Rat Model. J Med Food 2024; 27:627-635. [PMID: 38976324 DOI: 10.1089/jmf.2023.0022] [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] [Indexed: 07/09/2024] Open
Abstract
Type 2 diabetes (T2D) is a serious health problem, and its prevalence is expected to increase worldwide in the years ahead. Cruciferous vegetables such as Brassica oleracea var. capitata L. (green cabbage) and Raphanus sativus L. (radish) have therapeutic properties that can be used to support the treatment of T2D. This study evaluated the effect of B. oleracea (BAE) and R. sativus (RAE) aqueous extracts on zoometric parameters, glycemic profiles, and pancreas and liver in prediabetic rats induced by a high-sucrose diet (HSD). BAE and RAE were administered to male HSD-induced Wistar rats (n = 35) at 5 and 10 mg/kg doses for 5 weeks. Zoometric and biochemical changes were measured, and then the pancreas and liver histological preparations were analyzed to observe the protective effect. BAE decreased feed intake and weight gain. Both extracts decreased fasting glucose and insulin levels compared with control (not treated), although not significantly (P > .05). The extracts significantly (P < .05) reduced homeostatic model assessment for insulin resistance, homeostasis model assessment of β-cell function, and glucose intolerance, similar to metformin control. In addition, minor damage occurred in the pancreas and liver. The results indicated that BAE and RAE decreased weight gain, improved glucose regulation, and protected the pancreas and liver in HSD rats. Therefore, they have multiple therapeutical properties and may be helpful in the prevention of T2D.
Collapse
|
4
|
Yang KJ, Park H, Chang YK, Park CW, Kim SY, Hong YA. Xanthine oxidoreductase inhibition ameliorates high glucose-induced glomerular endothelial injury by activating AMPK through the purine salvage pathway. Sci Rep 2024; 14:11167. [PMID: 38750091 PMCID: PMC11096301 DOI: 10.1038/s41598-024-61436-1] [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: 03/02/2024] [Accepted: 05/06/2024] [Indexed: 05/18/2024] Open
Abstract
Xanthine oxidoreductase (XOR) contributes to reactive oxygen species production. We investigated the cytoprotective mechanisms of XOR inhibition against high glucose (HG)-induced glomerular endothelial injury, which involves activation of the AMP-activated protein kinase (AMPK). Human glomerular endothelial cells (GECs) exposed to HG were subjected to febuxostat treatment for 48 h and the expressions of AMPK and its associated signaling pathways were evaluated. HG-treated GECs were increased xanthine oxidase/xanthine dehydrogenase levels and decreased intracellular AMP/ATP ratio, and these effects were reversed by febuxostat treatment. Febuxostat enhanced the phosphorylation of AMPK, the activation of peroxisome proliferator-activated receptor (PPAR)-gamma coactivator (PGC)-1α and PPAR-α and suppressed the phosphorylation of forkhead box O (FoxO)3a in HG-treated GECs. Febuxostat also decreased nicotinamide adenine dinucleotide phosphate oxidase (Nox)1, Nox2, and Nox4 expressions; enhanced superoxide dismutase activity; and decreased malondialdehyde levels in HG-treated GECs. The knockdown of AMPK inhibited PGC-1α-FoxO3a signaling and negated the antioxidant effects of febuxostat in HG-treated GECs. Despite febuxostat administration, the knockdown of hypoxanthine phosphoribosyl transferase 1 (HPRT1) also inhibited AMPK-PGC-1α-FoxO3a in HG-treated GECs. XOR inhibition alleviates oxidative stress by activating AMPK-PGC-1α-FoxO3a signaling through the HPRT1-dependent purine salvage pathway in GECs exposed to HG conditions.
Collapse
Affiliation(s)
- Keum-Jin Yang
- Clinical Research Institute, Daejeon St. Mary's Hospital, Daejeon, Republic of Korea
| | - Hwajin Park
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoon-Kyung Chang
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Cheol Whee Park
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Suk Young Kim
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yu Ah Hong
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Hammad Uddin MK, Khan Sadiq MS, Ahmed A, Khan M, Maniar T, Mateen SM, Saba B, Kashif SM, Usman S, Najeeb S, Khurshid Z, Zafar MS. Applications of Metformin in Dentistry-A review. J Taibah Univ Med Sci 2023; 18:1299-1310. [PMID: 37275952 PMCID: PMC10239065 DOI: 10.1016/j.jtumed.2023.03.014] [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: 11/12/2022] [Revised: 03/10/2023] [Accepted: 03/30/2023] [Indexed: 06/07/2023] Open
Abstract
Metformin is a versatile drug with numerous medical uses. It is known primarily as an anti-hyperglycemic drug that has become the main oral blood-glucose-lowering medication for managing type 2 diabetes mellitus globally. Its use has been reported in a variety of oral conditions and dentistry in general. Recent clinical trials have indicated the effectiveness of adjunct topical application of metformin in improving the periodontal parameters of patients with diabetes and periodontitis. Additionally, studies have suggested that metformin stimulates odontogenic differentiation and mineral synthesis of stem cells in the tooth pulp. Metformin also stimulates osteoblast proliferation, decreases osteoclast activity and exerts regenerative effects on periodontal bone, thus making it a viable candidate for periodontal regeneration. Metformin monotherapy significantly enhances osseointegration of endosseous implants and has been reported to have anti-cancer effects on oral squamous cell carcinoma by impeding tumor progression. Animal studies have indicated that metformin improves orthodontic tooth movement and resists orthodontic appliance corrosion. This narrative review aims to provide a current summary of research highlighting the prospective uses of metformin in dentistry.
Collapse
Affiliation(s)
- Muhammad Khawaja Hammad Uddin
- Department of Science of Dental Materials, Dr. Ishrat-ul-Ebad Khan Institute of Oral Health Sciences, Dow University of Health Sciences, Karachi, Sindh, Pakistan
- School of Dental Care Professionals (SDCP), Dow University of Health Sciences Karachi, Sindh, Pakistan
| | - Muhammad Shahrukh Khan Sadiq
- Department of Oral Pathology, Bahria University Dental College, Bahria University Health Sciences Campus (Karachi) Karachi, Sindh, Pakistan
| | - Ashfaq Ahmed
- Department of Science of Dental Materials, Dr. Ishrat-ul-Ebad Khan Institute of Oral Health Sciences, Dow University of Health Sciences, Karachi, Sindh, Pakistan
- Department of Oral Pathology, Bahria University Dental College, Bahria University Health Sciences Campus (Karachi) Karachi, Sindh, Pakistan
| | - Mariam Khan
- Department of Science of Dental Materials, Dr. Ishrat-ul-Ebad Khan Institute of Oral Health Sciences, Dow University of Health Sciences, Karachi, Sindh, Pakistan
- Department of Oral Pathology, Bahria University Dental College, Bahria University Health Sciences Campus (Karachi) Karachi, Sindh, Pakistan
| | - Tooba Maniar
- Department of Science of Dental Materials, Dr. Ishrat-ul-Ebad Khan Institute of Oral Health Sciences, Dow University of Health Sciences, Karachi, Sindh, Pakistan
- Department of Oral Pathology, Bahria University Dental College, Bahria University Health Sciences Campus (Karachi) Karachi, Sindh, Pakistan
| | - Syeda Mamoona Mateen
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Sindh, Pakistan
| | - Bilquees Saba
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Sindh, Pakistan
- Department of Medicine, Ziauddin Medical College, Ziauddin University, Karachi, Sindh, Pakistan
| | - Syed Muhammad Kashif
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Sindh, Pakistan
- Department of General Medicine, Civil Hospital, Dow University of Health Sciences, Karachi, Sindh, Pakistan
| | - Shumaila Usman
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Sindh, Pakistan
- Department of Molecular Medicine, Ziauddin Medical College, Ziauddin University, Karachi, Sindh, Pakistan
| | - Shariq Najeeb
- Evidentia Dental Outcomes Research, Calgary, Alberta, Canada
- Schulich Dentistry, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C, Canada
| | - Zohaib Khurshid
- Department of Prosthodontics and Dental Implantology, King Faisal University, Hofuf, Al-Ahsa, Saudi Arabia
- Center of Excellence for Regenerative Dentistry, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Muhammad Sohail Zafar
- Department of Restorative Dentistry, College of Dentistry, Taibah University, Al Madina Al Munawara, 41311, Saudi Arabia
- Department of Dental Materials, Islamic International Dental College, Riphah International University, Islamabad 44000, Pakistan
| |
Collapse
|
6
|
Zheng Z, Wu L, Deng W, Yi K, Li Y. Polyphenol Composition, Antioxidant Capacity and Xanthine Oxidase Inhibition Mechanism of Furong Plum Fruits at Different Maturity Stages. Foods 2023; 12:4253. [PMID: 38231765 PMCID: PMC10705914 DOI: 10.3390/foods12234253] [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: 10/27/2023] [Revised: 11/17/2023] [Accepted: 11/22/2023] [Indexed: 01/19/2024] Open
Abstract
An experiment was conducted on the polyphenol content, flavonoid content, anthocyanin content, and antioxidant capacity of Furong plum (Prunus salicina Lindl. cv. "furong") at different maturity stages to determine the most suitable maturity stage. The inhibition of plum polyphenols on xanthine oxidase (XOD) was measured, and its kinetics were studied to reveal the inhibitory mechanism. The experimental results showed that the polyphenol, flavonoid and anthocyanin contents of plums at the ripe stage were the highest, reaching 320.46 mg GAE/100 g FW, 204.21 mg/100 g FW, and 66.24 mg/100 g FW, respectively, in comparison those of the plums at the immature and mid-ripe stages. The antioxidant capacity of the ripe plums was stronger than it was during the other stages of the plums growth. Among them, the total polyphenols of the ripe plums exhibited the strongest antioxidant capacity (IC50 values against DPPH and hydroxyl radicals were 28.19 ± 0.67 μg/mL and 198.16 ± 7.55 μg/mL, respectively), which was between the antioxidant capacity of the free polyphenols and bound polyphenols. The major phenolic monomer compounds of plum polyphenols were flavan-3-ols (epicatechin, catechin, proanthocyanidin, and procyanidin B2), flavonols (myricetin), and phenolic acids (chlorogenic acid, ferulic acid, and protocatechuic acid). Additionally, plum polyphenols exhibited a strong inhibitory effect on XOD, with an IC50 value of 77.64 μg/mL. The inhibition kinetics showed that plum polyphenols are mixed-type inhibitors that inhibit XOD activity and that the inhibition process is reversible. The calculated values of Ki and α were 16.53 mmol/L and 0.26, respectively.
Collapse
Affiliation(s)
- Zhipeng Zheng
- Institute of Food Science and Technology, Fujian Academy of Agricultural Sciences, Fuzhou 350003, China
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Li Wu
- Institute of Food Science and Technology, Fujian Academy of Agricultural Sciences, Fuzhou 350003, China
- Key Laboratory of Subtropical Characteristic Fruits, Vegetables and Edible Fungi Processing (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Fuzhou 350003, China
- Fujian Province Key Laboratory of Agricultural Products (Food) Processing Technology, Fuzhou 350003, China
| | - Wei Deng
- Institute of Food Science and Technology, Fujian Academy of Agricultural Sciences, Fuzhou 350003, China
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Kexin Yi
- Institute of Food Science and Technology, Fujian Academy of Agricultural Sciences, Fuzhou 350003, China
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yibin Li
- Institute of Food Science and Technology, Fujian Academy of Agricultural Sciences, Fuzhou 350003, China
- Key Laboratory of Subtropical Characteristic Fruits, Vegetables and Edible Fungi Processing (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Fuzhou 350003, China
- Fujian Province Key Laboratory of Agricultural Products (Food) Processing Technology, Fuzhou 350003, China
| |
Collapse
|
7
|
Yu W, Xie D, Yamamoto T, Koyama H, Cheng J. Mechanistic insights of soluble uric acid-induced insulin resistance: Insulin signaling and beyond. Rev Endocr Metab Disord 2023; 24:327-343. [PMID: 36715824 DOI: 10.1007/s11154-023-09787-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/10/2023] [Indexed: 01/31/2023]
Abstract
Hyperuricemia is a metabolic disease caused by purine nucleotide metabolism disorder. The prevalence of hyperuricemia is increasing worldwide, with a growing trend in the younger populations. Although numerous studies have indicated that hyperuricemia may be an independent risk factor for insulin resistance, the causal relationship between the two is controversial. There are few reviews, however, focusing on the relationship between uric acid (UA) and insulin resistance from experimental studies. In this review, we summarized the experimental models related to soluble UA-induced insulin resistance in pancreas and peripheral tissues, including skeletal muscles, adipose tissue, liver, heart/cardiomyocytes, vascular endothelial cells and macrophages. In addition, we summarized the research advances about the key mechanism of UA-induced insulin resistance. Moreover, we attempt to identify novel targets for the treatment of hyperuricemia-related insulin resistance. Lastly, we hope that the present review will encourage further researches to solve the chicken-and-egg dilemma between UA and insulin resistance, and provide strategies for the pathogenesis and treatment of hyperuricemia related metabolic diseases.
Collapse
Affiliation(s)
- Wei Yu
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - De Xie
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Tetsuya Yamamoto
- Health Evaluation Center, Osaka Gyoumeikan Hospital, Osaka, Japan
| | - Hidenori Koyama
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Jidong Cheng
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo Medical University, Nishinomiya, Hyogo, Japan.
- Xiamen Key Laboratory of Translational Medicine for Nucleic Acid Metabolism and Regulation, Xiamen, Fujian, China.
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China.
| |
Collapse
|
8
|
He F, Wang M, Zhao H, Xie D, Lv J, Liu W, Yu W, Wang Q, Chen B, Xu C, Yamamoto T, Koyama H, Cheng J. Autophagy protects against high uric acid-induced hepatic insulin resistance. Mol Cell Endocrinol 2022; 547:111599. [PMID: 35181437 DOI: 10.1016/j.mce.2022.111599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 10/19/2022]
Abstract
Uric acid (UA), the end-product of purine metabolism, is closely related to hepatic insulin resistance (IR). Autophagy is a conserved intracellular degradation process maintaining cellular homeostasis. Autophagy plays a protective role in obesity-related hepatic IR, but whether it occurs in high uric acid (HUA)-induced hepatic IR is unclear. In this study, spontaneously elevated UA level induced hepatic IR and facilitated hepatic autophagy degradation in uricase knockout (Uox-/-) mice. In vitro, HepG2 cells stimulated with HUA medium showed decreased glucose uptake and inhibition of insulin signaling pathways, concomitant with activation of autophagy, as manifested by increased conversion of LC3B-I to -II. Rapamycin, the autophagy activator, alleviated but the autophagy inhibitor trimethyl adenine (3-MA) aggravated HUA-induced IR in HepG2 cells. Similarly, rapamycin ameliorated and 3-MA worsened HUA-induced blood glucose level and hepatic IR in Uox-/- mice. Mechanistically, HUA enhanced AMPKα phosphorylation (p-AMPKα) and inhibited mammalian target of rapamycin phosphorylation (p-mTOR) in HepG2 cells. The levels of p-AMPKα and LC3B-II/I were downregulated in HepG2 cells transfected with small interfering RNA (siRNA) against AMPKα, which suggests that the AMPKα-mTOR pathway was involved in HUA-induced autophagy. Antioxidant N-acetyl-L-cysteine reversed elevated reactive oxygen species levels induced by HUA in HepG2 cells, and AMPKα level was also inhibited, which suggests that AMPKα activation may be derived from reactive oxygen species. Collectively, these findings demonstrate that HUA increased hepatic autophagy, and autophagy activation plays a protective role in hepatic IR, which may suggest a potential therapeutic target for hepatic IR derived from HUA.
Collapse
Affiliation(s)
- Furong He
- Department of Endocrinology, Xiang'an Hospital of Xiamen University. Xiamen, Fujian, China
| | - Mei Wang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, China
| | - Hairong Zhao
- Department of Endocrinology, Xiang'an Hospital of Xiamen University. Xiamen, Fujian, China
| | - De Xie
- Department of Endocrinology, Xiang'an Hospital of Xiamen University. Xiamen, Fujian, China
| | - Jiaming Lv
- Department of Endocrinology, Xiang'an Hospital of Xiamen University. Xiamen, Fujian, China
| | - Weidong Liu
- Department of Endocrinology, Xiang'an Hospital of Xiamen University. Xiamen, Fujian, China
| | - Wei Yu
- Department of Endocrinology, Xiang'an Hospital of Xiamen University. Xiamen, Fujian, China
| | - Qiang Wang
- Department of Endocrinology, Xiang'an Hospital of Xiamen University. Xiamen, Fujian, China
| | - Binyang Chen
- Department of Endocrinology, Xiang'an Hospital of Xiamen University. Xiamen, Fujian, China
| | - Chenxi Xu
- Department of Endocrinology, Xiang'an Hospital of Xiamen University. Xiamen, Fujian, China
| | - Tetsuya Yamamoto
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Hidenori Koyama
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Jidong Cheng
- Department of Endocrinology, Xiang'an Hospital of Xiamen University. Xiamen, Fujian, China; Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan.
| |
Collapse
|
9
|
Metformin: Expanding the Scope of Application-Starting Earlier than Yesterday, Canceling Later. Int J Mol Sci 2022; 23:ijms23042363. [PMID: 35216477 PMCID: PMC8875586 DOI: 10.3390/ijms23042363] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/29/2022] [Accepted: 02/18/2022] [Indexed: 12/15/2022] Open
Abstract
Today the area of application of metformin is expanding, and a wealth of data point to its benefits in people without carbohydrate metabolism disorders. Already in the population of people leading an unhealthy lifestyle, before the formation of obesity and prediabetes metformin smooths out the adverse effects of a high-fat diet. Being prescribed at this stage, metformin will probably be able to, if not prevent, then significantly reduce the progression of all subsequent metabolic changes. To a large extent, this review will discuss the proofs of the evidence for this. Another recent important change is a removal of a number of restrictions on its use in patients with heart failure, acute coronary syndrome and chronic kidney disease. We will discuss the reasons for these changes and present a new perspective on the role of increasing lactate in metformin therapy.
Collapse
|
10
|
Zhao H, Lu Z, Lu Y. The potential of probiotics in the amelioration of hyperuricemia. Food Funct 2022; 13:2394-2414. [PMID: 35156670 DOI: 10.1039/d1fo03206b] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hyperuricemia is a common disease caused by metabolic disorders or the excessive intake of high-purine foods. Persistent hyperuricemia in extreme cases induces gout, and asymptomatic hyperuricemia is probably linked to other metabolic diseases, such as hypertension. The typical damage caused by asymptomatic hyperuricemia includes inflammation, oxidative stress and gut dysbiosis. Probiotics have broad potential applications as food additives, not as drug therapies, in the amelioration of hyperuricemia. In this review, we describe novel methods for potential hyperuricemia amelioration with probiotics. The pathways through which probiotics may ameliorate hyperuricemia are discussed, including the decrease in uric acid production through purine assimilation and XOD (xanthine oxidase) inhibition as well as enhanced excretion of uric acid production by promoting ABCG2 (ATP binding cassette subfamily G member 2) activity, respectively. Three possible probiotic-related therapeutic pathways for alleviating the syndrome of hyperuricemia are also summarized. The first mechanism is to alleviate the oxidation and inflammation induced by hyperuricemia through the inhibition of NLRP3 inflammasome, the second is to restore damaged intestinal epithelium barriers and prevent gut microbiota dysbiosis, and the third is to enhance the innate immune system by increasing the secretion of immunoglobulin A (sIgA) to resist the stimulus by hyperuricemia. We propose that future research should focus on superior strain resource isolation and insight into the cause-effect mechanisms of probiotics for hyperuricemia amelioration. The safety and effects of the application of probiotics in clinical use also need verification.
Collapse
Affiliation(s)
- Hongyuan Zhao
- College of Food Science & Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Zhaoxin Lu
- College of Food Science & Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Yingjian Lu
- College of Food Science & Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China.
| |
Collapse
|
11
|
Zhao H, Lu J, He F, Wang M, Yan Y, Chen B, Xie D, Xu C, Wang Q, Liu W, Yu W, Xi Y, Yu L, Yamamoto T, Koyama H, Wang W, Zhang C, Cheng J. Hyperuricemia contributes to glucose intolerance of hepatic inflammatory macrophages and impairs the insulin signaling pathway via IRS2-proteasome degradation. Front Immunol 2022; 13:931087. [PMID: 36177037 PMCID: PMC9513153 DOI: 10.3389/fimmu.2022.931087] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/24/2022] [Indexed: 02/05/2023] Open
Abstract
AIM Numerous reports have demonstrated the key importance of macrophage-elicited metabolic inflammation in insulin resistance (IR). Our previous studies confirmed that hyperuricemia or high uric acid (HUA) treatment induced an IR state in several peripheral tissues to promote the development of type 2 diabetes mellitus (T2DM). However, the effect of HUA on glucose uptake and the insulin sensitivity of macrophages and its mechanism is unclear. METHODS To assess systemic IR, we generated hyperuricemic mice by urate oxidase knockout (UOX-KO). Then, glucose/insulin tolerance, the tissue uptake of 18F-fluorodeoxyglucose, body composition, and energy balance were assessed. Glucose uptake of circulating infiltrated macrophages in the liver was evaluated by glucose transporter type 4 (GLUT-4) staining. Insulin sensitivity and the insulin signaling pathway of macrophages were demonstrated using the 2-NBDG kit, immunoblotting, and immunofluorescence assays. The immunoprecipitation assay and LC-MS analysis were used to determine insulin receptor substrate 2 (IRS2) levels and its interacting protein enrichment under HUA conditions. RESULTS Compared to WT mice (10 weeks old), serum uric acid levels were higher in UOX-KO mice (WT, 182.3 ± 5.091 μM versus KO, 421.9 ± 45.47 μM). Hyperuricemic mice with metabolic disorders and systemic IR showed inflammatory macrophage recruitment and increased levels of circulating proinflammatory cytokines. HUA inhibited the nuclear translocation of GLUT-4 in hepatic macrophages, restrained insulin-induced glucose uptake and glucose tolerance, and blocked insulin IRS2/PI3K/AKT signaling. Meanwhile, HUA mediated the IRS2 protein degradation pathway and activated AMPK/mTOR in macrophages. LC-MS analysis showed that ubiquitination degradation could be involved in IRS2 and its interacting proteins to contribute to IR under HUA conditions. CONCLUSION The data suggest that HUA-induced glucose intolerance in hepatic macrophages contributed to insulin resistance and impaired the insulin signaling pathway via IRS2-proteasome degradation.
Collapse
Affiliation(s)
- Hairong Zhao
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical Research and Development (R&D), College of Pharmacy, Dali University, Dali, China
| | - Jiaming Lu
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Furong He
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Mei Wang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical Research and Development (R&D), College of Pharmacy, Dali University, Dali, China
| | - Yunbo Yan
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Binyang Chen
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - De Xie
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Chenxi Xu
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Qiang Wang
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Weidong Liu
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Wei Yu
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Yuemei Xi
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Linqian Yu
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Tetsuya Yamamoto
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Hidenori Koyama
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Wei Wang
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Chenggui Zhang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical Research and Development (R&D), College of Pharmacy, Dali University, Dali, China
- *Correspondence: Chenggui Zhang, ; Jidong Cheng,
| | - Jidong Cheng
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Japan
- *Correspondence: Chenggui Zhang, ; Jidong Cheng,
| |
Collapse
|
12
|
Yu W, Chen C, Zhuang W, Wang W, Liu W, Zhao H, Lv J, Xie D, Wang Q, He F, Xu C, Chen B, Yamamoto T, Koyama H, Cheng J. Silencing TXNIP ameliorates high uric acid-induced insulin resistance via the IRS2/AKT and Nrf2/HO-1 pathways in macrophages. Free Radic Biol Med 2022; 178:42-53. [PMID: 34848368 DOI: 10.1016/j.freeradbiomed.2021.11.034] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 02/08/2023]
Abstract
Insulin resistance (IR) promotes atherosclerosis and increases the risk of diabetes and cardiovascular diseases. Our previous studies have demonstrated that high uric acid (HUA) increased oxidative stress, leading to IR in cardiomyocytes and pancreatic β cells. However, whether HUA can induce IR in monocytes/macrophages, which play critical roles in all stages of atherosclerosis, is unclear. Recent findings revealed that thioredoxin-interacting protein (TXNIP) negatively regulates insulin signaling; however, the roles and mechanisms of TXNIP in HUA-induced IR remain unclear. Therefore, in this study, we investigated the function of TXNIP in macrophages treated with UA. Transcriptomic profiling revealed TXNIP as one of the most upregulated genes, and subsequent RT-PCR and Western blot analyses confirmed that TXNIP was upregulated by HUA. HUA treatment significantly increased mitochondrial reactive oxygen species (MtROS) levels and decreased insulin-stimulated glucose uptake. Silencing TXNIP by RNA interference significantly diminished HUA-induced oxidative stress and IR. Mechanistically, silencing TXNIP reversed the inhibition of the phosphorylation of insulin receptor substrate 2 (IRS2)/protein kinase B (AKT) pathway induced by HUA. Additional study revealed that HUA induced the activation of the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) signaling pathway, but silencing TXNIP abolished it. Moreover, Nrf2 inhibitor (ML385) ameliorated HUA-induced IR independent of IRS2/AKT signaling. Probenecid, a well-known UA-lowering drug, significantly suppressed the activation of TXNIP and Nrf2/HO-1 signaling. Furthermore, RNA-seq revealed that activation of the TXNIP-related redox pathway may be a key regulator in patients with asymptomatic hyperuricemia. These data suggest that silencing TXNIP could ameliorate HUA-induced IR via the IRS2/AKT and Nrf2/HO-1 pathways in macrophages. Additionally, TXNIP might be a promising therapeutic target for preventing and treating oxidative stress and IR induced by HUA.
Collapse
Affiliation(s)
- Wei Yu
- Department of Internal Medicine, Xiang'an Hospital of Xiamen University, School of Medicine Xiamen University, Xiamen, China
| | - Chunjuan Chen
- Department of Cardiology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Wanling Zhuang
- Department of Hematology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Wei Wang
- Department of Internal Medicine, Xiang'an Hospital of Xiamen University, School of Medicine Xiamen University, Xiamen, China
| | - Weidong Liu
- Department of Internal Medicine, Xiang'an Hospital of Xiamen University, School of Medicine Xiamen University, Xiamen, China
| | - Hairong Zhao
- Department of Internal Medicine, Xiang'an Hospital of Xiamen University, School of Medicine Xiamen University, Xiamen, China
| | - Jiaming Lv
- Department of Internal Medicine, Xiang'an Hospital of Xiamen University, School of Medicine Xiamen University, Xiamen, China
| | - De Xie
- Department of Internal Medicine, Xiang'an Hospital of Xiamen University, School of Medicine Xiamen University, Xiamen, China
| | - Qiang Wang
- Department of Internal Medicine, Xiang'an Hospital of Xiamen University, School of Medicine Xiamen University, Xiamen, China
| | - Furong He
- Department of Internal Medicine, Xiang'an Hospital of Xiamen University, School of Medicine Xiamen University, Xiamen, China
| | - Chenxi Xu
- Department of Internal Medicine, Xiang'an Hospital of Xiamen University, School of Medicine Xiamen University, Xiamen, China
| | - Bingyang Chen
- Department of Internal Medicine, Xiang'an Hospital of Xiamen University, School of Medicine Xiamen University, Xiamen, China
| | - Tetsuya Yamamoto
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Hidenori Koyama
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Jidong Cheng
- Department of Internal Medicine, Xiang'an Hospital of Xiamen University, School of Medicine Xiamen University, Xiamen, China
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
- Xiamen Key Laboratory of Translational Medicine for Nucleic Acid Metabolism and Regulation, Xiamen, Fujian, China
| |
Collapse
|
13
|
Uuh-Narvaez JJ, Segura-Campos MR. Cabbage (Brassica oleracea var. capitata): A food with functional properties aimed to type 2 diabetes prevention and management. J Food Sci 2021; 86:4775-4798. [PMID: 34658044 DOI: 10.1111/1750-3841.15939] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/08/2021] [Accepted: 09/13/2021] [Indexed: 12/27/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is increasing the prevalence worldwide at an alarming rate, becoming a serious public health problem that mainly affects developing countries. Functional food research is currently of great interest because it contributes to developing nutritional therapy strategies for T2DM prevention and treatment. Bioactive compounds identified in some plant foods contribute to human health by mechanisms of action that exert biological effects on metabolic pathways involved in the development of T2DM. Hence, vegetables with high bioactive compounds content may be a source of functional value for the control of T2DM. Cabbages varieties (Brassica oleracea var. capitata) such as green (GCB), white (WCB), and red (RCB) are foods consumed (raw or cooked) and cultivated in different regions of the world. Scientific evidence shows that cabbage has multi-target effects on glucose homeostatic regulation due to its high content of bioactive compounds. It has also been shown to decrease damage to organs affected by T2DM complications, such as the liver and kidney. Additionally, it could contribute as a preventive by attenuating problems underlying the development of T2DM as oxidative stress and obesity. This review highlights the functional properties of cabbage varieties involved in glucose regulation and the main mechanisms of the action exerted by their bioactive compounds. In conclusion, cabbage is a valuable food that can be employed as part of nutritional therapy or functional ingredient aimed at the prevention and treatment of T2DM.
Collapse
|
14
|
Bahadoran Z, Mirmiran P, Kashfi K, Ghasemi A. Hyperuricemia-induced endothelial insulin resistance: the nitric oxide connection. Pflugers Arch 2021; 474:83-98. [PMID: 34313822 DOI: 10.1007/s00424-021-02606-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 05/12/2021] [Accepted: 07/08/2021] [Indexed: 12/15/2022]
Abstract
Hyperuricemia, defined as elevated serum concentrations of uric acid (UA) above 416 µmol L-1, is related to the development of cardiometabolic disorders, probably via induction of endothelial dysfunction. Hyperuricemia causes endothelial dysfunction via induction of cell apoptosis, oxidative stress, and inflammation; however, it's interfering with insulin signaling and decreased endothelial nitric oxide (NO) availability, resulting in the development of endothelial insulin resistance, which seems to be a major underlying mechanism for hyperuricemia-induced endothelial dysfunction. Here, we elaborate on how hyperuricemia induces endothelial insulin resistance through the disruption of insulin-stimulated endothelial NO synthesis. High UA concentrations decrease insulin-induced NO synthesis within the endothelial cells by interfering with insulin signaling at either the receptor or post-receptor levels (i.e., proximal and distal steps). At the proximal post-receptor level, UA impairs the function of the insulin receptor substrate (IRS) and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) in the insulin signaling pathway. At the distal level, high UA concentrations impair endothelial NO synthase (eNOS)-NO system by decreasing eNOS expression and activity as well as by direct inactivation of NO. Clinically, UA-induced endothelial insulin resistance is translated into impaired endothelial function, impaired NO-dependent vasodilation, and the development of systemic insulin resistance. UA-lowering drugs may improve endothelial function in subjects with hyperuricemia.
Collapse
Affiliation(s)
- Zahra Bahadoran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, 10031, USA.,Graduate Program in Biology, City University of New York Graduate Center, New York, NY, 10016, USA
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24, Parvaneh Street, P.O. Box: 19395-4763, VelenjakTehran, Iran.
| |
Collapse
|
15
|
Sánchez-Alonso P, Griera M, García-Marín J, Rodríguez-Puyol M, Alajarín R, Vaquero JJ, Rodríguez-Puyol D. Pyrrolo[1,2-a]quinoxal-5-inium salts and 4,5-dihydropyrrolo[1,2-a]quinoxalines: Synthesis, activity and computational docking for protein tyrosine phosphatase 1B. Bioorg Med Chem 2021; 44:116295. [PMID: 34246920 DOI: 10.1016/j.bmc.2021.116295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/18/2021] [Accepted: 06/19/2021] [Indexed: 11/17/2022]
Abstract
Protein tyrosine phosphatase (PTP1B) is an interesting therapeutical target for diabetes, obesity, heart disease and cancer. As such, inhibition of PTP1B using orally administered drugs is still being pursued by academia and pharmaceutical companies. The failure of catalytic-site inhibitors led to the focus in this field being switched to allosteric inhibitors. To date, the non-competitive inhibitors that have reached clinical trials target the site formed by the α3/α6/α7 tunnel or the site found in a disordered C-terminal non-catalytic segment. Herein, pyrrolo[1,2-a]quinoxal-5-inium salts and 4,5-dihydropyrrolo[1,2-a]quinoxalines are synthesized from pyrrolo[1,2-a]quinoxalines by alkylation and reduction, respectively. These compounds showed no toxicity in HepG2 cells and exhibited inhibitory activity against PTP1B, with inhibition percentages of between 37% and 53% at 1 μM and activities (IC50) of between 0.25 and 1.90 μM. The inhibitory activity against T-cell protein tyrosine phosphatase (TC-TPT) was also assayed, with 4,5-dihydropyrrolo[1,2-a]quinoxalines being found to be slightly more active and selective. Compounds from the two series behave as insulin mimetics since they exhibit enhancement of glucose uptake in C2C12 cells. Computational docking studies provide information about the putative binding mode for both series and the preference for the α3/α6/α7 allosteric tunnel.
Collapse
Affiliation(s)
- Patricia Sánchez-Alonso
- Departamento de Química Orgánica y Química Inorgánica, Universidad de Alcalá, 28805 Alcalá de Henares, Madrid, Spain
| | | | - Javier García-Marín
- Departamento de Química Orgánica y Química Inorgánica, Universidad de Alcalá, 28805 Alcalá de Henares, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Ctra. Colmenar Viejo, km. 9100, 28034 Madrid, Spain; Instituto de Investigación Química Andrés M. del Río (IQAR), Facultad de Farmacia, Universidad de Alcalá, 28805 Alcalá de Henares, Madrid, Spain
| | - Manuel Rodríguez-Puyol
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Ctra. Colmenar Viejo, km. 9100, 28034 Madrid, Spain; Departamento de Biología de Sistemas, Universidad de Alcalá, 28805 Alcalá de Henares, Madrid, Spain; Fundación Renal Iñigo Álvarez de Toledo (FRIAT) y REDinREN del Instituto de Salud Carlos III, Madrid, Spain
| | - Ramón Alajarín
- Departamento de Química Orgánica y Química Inorgánica, Universidad de Alcalá, 28805 Alcalá de Henares, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Ctra. Colmenar Viejo, km. 9100, 28034 Madrid, Spain; Instituto de Investigación Química Andrés M. del Río (IQAR), Facultad de Farmacia, Universidad de Alcalá, 28805 Alcalá de Henares, Madrid, Spain.
| | - Juan J Vaquero
- Departamento de Química Orgánica y Química Inorgánica, Universidad de Alcalá, 28805 Alcalá de Henares, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Ctra. Colmenar Viejo, km. 9100, 28034 Madrid, Spain; Instituto de Investigación Química Andrés M. del Río (IQAR), Facultad de Farmacia, Universidad de Alcalá, 28805 Alcalá de Henares, Madrid, Spain
| | - Diego Rodríguez-Puyol
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Ctra. Colmenar Viejo, km. 9100, 28034 Madrid, Spain; Departamento de Biología de Sistemas, Universidad de Alcalá, 28805 Alcalá de Henares, Madrid, Spain; Fundación Renal Iñigo Álvarez de Toledo (FRIAT) y REDinREN del Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
16
|
Jiao Z, Chen Y, Xie Y, Li Y, Li Z. Metformin protects against insulin resistance induced by high uric acid in cardiomyocytes via AMPK signalling pathways in vitro and in vivo. J Cell Mol Med 2021; 25:6733-6745. [PMID: 34053175 PMCID: PMC8278091 DOI: 10.1111/jcmm.16677] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/04/2021] [Accepted: 05/08/2021] [Indexed: 02/05/2023] Open
Abstract
High uric acid (HUA) is associated with insulin resistance (IR) in cardiomyocytes. We investigated whether metformin protects against HUA-induced IR in cardiomyocytes. We exposed primary cardiomyocytes to HUA, and cellular glucose uptake was quantified by measuring the uptake of 2-NBDG, a fluorescent glucose analog. Western blot was used to examine the levels of signalling protein. Membrane of glucose transporter type 4 (GLUT4) was analysed by immunofluorescence. We monitored the impact of metformin on HUA-induced IR and in myocardial tissue of an acute hyperuricaemia mouse model established by potassium oxonate treatment. Treatment with metformin protected against HUA-reduced glucose uptake induced by insulin in cardiomyocytes. HUA directly inhibited the phosphorylation of Akt and the translocation of GLUT4 induced by insulin, which was blocked by metformin. Metformin promoted phosphorylation of AMP-activated protein kinase (AMPK) and restored the insulin-stimulated glucose uptake in HUA-induced IR cardiomyocytes. As a result of these effects, in a mouse model of acute hyperuricaemia, metformin improved insulin tolerance and glucose tolerance, accompanied by increased AMPK phosphorylation, Akt phosphorylation and translocation of GLUT4 in myocardial tissues. As expected, AICAR, another AMPK activator, had similar effects to metformin, demonstrating the important role of AMPK activation in protecting against IR induced by HUA in cardiomyocytes. Metformin protects against IR induced by HUA in cardiomyocytes and improves insulin tolerance and glucose tolerance in an acute hyperuricaemic mouse model, along with the activation of AMPK. Consequently, metformin may be an important potential new treatment strategy for hyperuricaemia-related cardiovascular disease.
Collapse
Affiliation(s)
- Zhenyu Jiao
- Department of CardiologyBeijing Chaoyang HospitalBeijingChina
- Department of CardiologySecond Affiliated Hospital of Shantou University Medical CollegeShantou, GuangdongChina
| | - Yingqun Chen
- Department of CardiologySecond Affiliated Hospital of Shantou University Medical CollegeShantou, GuangdongChina
- Department of Intensive Care UnitPeking University Shenzhen HospitalShenzhenChina
| | - Yang Xie
- Department of CardiologySecond Affiliated Hospital of Shantou University Medical CollegeShantou, GuangdongChina
| | - Yanbing Li
- Department of CardiologyBeijing Chaoyang HospitalBeijingChina
- Department of CardiologyBeijing You An HospitalBeijingChina
| | - Zhi Li
- Department of CardiologySecond Affiliated Hospital of Shantou University Medical CollegeShantou, GuangdongChina
| |
Collapse
|
17
|
Zhang R, Zhou X, Sheng Q, Zhang Q, Xie T, Xu C, Zou Z, Dong J, Liao L. Gliquidone ameliorates hepatic insulin resistance in streptozotocin-induced diabetic Sur1 -/- rats. Eur J Pharmacol 2021; 906:174221. [PMID: 34081903 DOI: 10.1016/j.ejphar.2021.174221] [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: 12/26/2020] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 11/17/2022]
Abstract
Gliquidone was suggested to exert hypoglycemic effect through enhancing hepatic insulin sensitivity. However, inadequate in vivo evidences make this statement controversial. The aim of the present study was to clarify the insulin-sensitizer role of gliquidone in liver and muscle, so as to confirm its extra-pancreatic effects in vivo. TALEN technique was used to create Sur1 knockout (Sur1-/-) rats. Diabetic Sur1-/- rat models were established by high-fat diet combined with streptozotocin, and which were randomly divided into three groups: gliquidone, metformin and saline, treated for 8 weeks. Fasting blood glucose (FBG) and body mass were tested each week. IPGTT, IPITT and hyperinsulinemic-euglycemic clamp tests were used to evaluate glucose tolerance and insulin sensitivity, respectively. Key mediators of glucose metabolism in liver and skeletal muscle and the activity of AKT and AMPK in these tissues were further analyzed. We found that gliquidone decreased FBG and increased insulin sensitivity without increasing insulin secretion in diabetic Sur1-/- rats. Further exploration implied that gliquidone mainly increased hepatic glycogen storage and decreased gluconeogenesis, which were accompanied with activation of AKT, but not enhanced muscle GLUT4 expression. However, both these effects were still weaker than that of metformin. These results suggested that gliquidone could exerts an extra-pancreatic hypoglycemic effect by improving insulin sensitivity, which might be largely attributes to its additional insulin sensitizer role in hepatic glucose metabolism.
Collapse
Affiliation(s)
- Rui Zhang
- Division of Endocrinology, Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Xiaojun Zhou
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Laboratory of Endocrinology, Jinan, 250014, China; Division of Endocrinology, Department of Internal Medicine, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250014, China
| | - Qiqi Sheng
- Division of Endocrinology, Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Qian Zhang
- Department of Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Tianyue Xie
- Division of Endocrinology, Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Chunmei Xu
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Laboratory of Endocrinology, Jinan, 250014, China; Division of Endocrinology, Department of Internal Medicine, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250014, China
| | - Zhiwei Zou
- Division of Endocrinology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264117, China
| | - Jianjun Dong
- Division of Endocrinology, Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Lin Liao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Laboratory of Endocrinology, Jinan, 250014, China; Division of Endocrinology, Department of Internal Medicine, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250014, China.
| |
Collapse
|
18
|
Hu Y, Zhao H, Lu J, Xie D, Wang Q, Huang T, Xin H, Hisatome I, Yamamoto T, Wang W, Cheng J. High uric acid promotes dysfunction in pancreatic β cells by blocking IRS2/AKT signalling. Mol Cell Endocrinol 2021; 520:111070. [PMID: 33127482 DOI: 10.1016/j.mce.2020.111070] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 10/24/2020] [Accepted: 10/26/2020] [Indexed: 02/05/2023]
Abstract
Hyperuricaemia is a disorder of purine metabolism. Elevated serum uric acid is strongly associated with many diseases, including gout, abdominal obesity, insulin resistance, and cardiovascular and kidney disease. Our previous studies showed that high uric acid (HUA) induced insulin resistance in several peripheral organs, including the liver, myocardium and adipose tissue. However, whether HUA directly induces insulin resistance of pancreatic β cells, the only source of insulin in the body and also a sensitive insulin target, is unknown. In this study, pancreatic β cells pretreated with HUA showed impaired insulin expression/secretion, glucose uptake and the glycolytic pathway. RNA-seq revealed that HUA affected the biological processes of INS-1 cells broadly, including oxidoreduction coenzyme metabolic process, pyruvate metabolic process, and glycolytic process. In addition, HUA reduced mitochondrial membrane potential and increased the production of reactive oxygen species(ROS) in INS-1 cells. INS-1 cells pretreated with probenecid, an organic anion transporter inhibitor, protected INS-1 cells against HUA-induced insulin secretion decrease, Pretreatment with N-acetyl-L-cysteine(NAC), a globally used antioxidant, recovered HUA-decreased insulin secretion and glucose uptake by pancreatic β cells. Insulin-like growth factor 1 (IGF-1), the phosphatidylinositol 3-kinase (PI3K) activator, rescues HUA-decreased insulin secretion by re-activating AKT phosphorylation. Thus, HUA induce insulin resistance, impaired insulin secretion and glycolytic pathway of pancreatic ꞵ cell through IRS2/AKT pathway.
Collapse
Affiliation(s)
- Yaqiu Hu
- Department of Internal Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Hairong Zhao
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Jiaming Lu
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - De Xie
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Qiang Wang
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Tianliang Huang
- Department of Internal Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Hancheng Xin
- Department of Internal Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Ichiro Hisatome
- Division of Regenerative Medicine and Therapeutics, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Sciences, Tottori University, Yonago, Japan
| | - Tetsuya Yamamoto
- Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Wei Wang
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China.
| | - Jidong Cheng
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
19
|
García‐Marín J, Griera M, Sánchez‐Alonso P, Di Geronimo B, Mendicuti F, Rodríguez‐Puyol M, Alajarín R, Pascual‐Teresa B, Vaquero JJ, Rodríguez‐Puyol D. Pyrrolo[1,2‐
a
]quinoxalines: Insulin Mimetics that Exhibit Potent and Selective Inhibition against Protein Tyrosine Phosphatase 1B. ChemMedChem 2020; 15:1788-1801. [DOI: 10.1002/cmdc.202000446] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 07/29/2020] [Indexed: 12/29/2022]
Affiliation(s)
- Javier García‐Marín
- Departamento de Química Orgánica y Química Inorgánica Universidad de Alcalá 28805 Alcalá de Henares Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) Ctra. Colmenar Viejo, km. 9100 28034 Madrid Spain
- Instituto de Investigación Química Andrés M. del Río Facultad de Farmacia Universidad de Alcalá 28805 Alcalá de Henares Spain
| | - Mercedes Griera
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) Ctra. Colmenar Viejo, km. 9100 28034 Madrid Spain
- Departamento de Biología de Sistemas Universidad de Alcalá 28805 Alcalá de Henares Spain
| | - Patricia Sánchez‐Alonso
- Departamento de Química Orgánica y Química Inorgánica Universidad de Alcalá 28805 Alcalá de Henares Spain
| | - Bruno Di Geronimo
- Departamento de Química y Bioquímica Facultad de Farmacia Universidad San Pablo CEU 28925 Alcorcón Spain
| | - Francisco Mendicuti
- Departamento de Química Analítica Química Física e Ingeniería Química Universidad de Alcalá 28805 Alcalá de Henares Spain
- Instituto de Investigación Química Andrés M. del Río Facultad de Farmacia Universidad de Alcalá 28805 Alcalá de Henares Spain
| | - Manuel Rodríguez‐Puyol
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) Ctra. Colmenar Viejo, km. 9100 28034 Madrid Spain
- Departamento de Biología de Sistemas Universidad de Alcalá 28805 Alcalá de Henares Spain
| | - Ramón Alajarín
- Departamento de Química Orgánica y Química Inorgánica Universidad de Alcalá 28805 Alcalá de Henares Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) Ctra. Colmenar Viejo, km. 9100 28034 Madrid Spain
- Instituto de Investigación Química Andrés M. del Río Facultad de Farmacia Universidad de Alcalá 28805 Alcalá de Henares Spain
| | - Beatriz Pascual‐Teresa
- Departamento de Química y Bioquímica Facultad de Farmacia Universidad San Pablo CEU 28925 Alcorcón Spain
| | - Juan J. Vaquero
- Departamento de Química Orgánica y Química Inorgánica Universidad de Alcalá 28805 Alcalá de Henares Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) Ctra. Colmenar Viejo, km. 9100 28034 Madrid Spain
- Instituto de Investigación Química Andrés M. del Río Facultad de Farmacia Universidad de Alcalá 28805 Alcalá de Henares Spain
| | - Diego Rodríguez‐Puyol
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) Ctra. Colmenar Viejo, km. 9100 28034 Madrid Spain
- Departamento de Biología de Sistemas Universidad de Alcalá 28805 Alcalá de Henares Spain
| |
Collapse
|
20
|
Mudali D, Jeevanandam J, Danquah MK. Probing the characteristics and biofunctional effects of disease-affected cells and drug response via machine learning applications. Crit Rev Biotechnol 2020; 40:951-977. [PMID: 32633615 DOI: 10.1080/07388551.2020.1789062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Drug-induced transformations in disease characteristics at the cellular and molecular level offers the opportunity to predict and evaluate the efficacy of pharmaceutical ingredients whilst enabling the optimal design of new and improved drugs with enhanced pharmacokinetics and pharmacodynamics. Machine learning is a promising in-silico tool used to simulate cells with specific disease properties and to determine their response toward drug uptake. Differences in the properties of normal and infected cells, including biophysical, biochemical and physiological characteristics, plays a key role in developing fundamental cellular probing platforms for machine learning applications. Cellular features can be extracted periodically from both the drug treated, infected, and normal cells via image segmentations in order to probe dynamic differences in cell behavior. Cellular segmentation can be evaluated to reflect the levels of drug effect on a distinct cell or group of cells via probability scoring. This article provides an account for the use of machine learning methods to probe differences in the biophysical, biochemical and physiological characteristics of infected cells in response to pharmacokinetics uptake of drug ingredients for application in cancer, diabetes and neurodegenerative disease therapies.
Collapse
Affiliation(s)
- Deborah Mudali
- Department of Computer Science, University of Tennessee, Chattanooga, TN, USA
| | - Jaison Jeevanandam
- Department of Chemical Engineering, Faculty of Engineering and Science, Curtin University, Miri, Malaysia
| | - Michael K Danquah
- Chemical Engineering Department, University of Tennessee, Chattanooga, TN, USA
| |
Collapse
|
21
|
Joosten LAB, Crişan TO, Bjornstad P, Johnson RJ. Asymptomatic hyperuricaemia: a silent activator of the innate immune system. Nat Rev Rheumatol 2020; 16:75-86. [PMID: 31822862 PMCID: PMC7075706 DOI: 10.1038/s41584-019-0334-3] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2019] [Indexed: 12/22/2022]
Abstract
Asymptomatic hyperuricaemia affects ~20% of the general population in the USA, with variable rates in other countries. Historically, asymptomatic hyperuricaemia was considered a benign laboratory finding with little clinical importance in the absence of gout or kidney stones. Yet, increasing evidence suggests that asymptomatic hyperuricaemia can predict the development of hypertension, obesity, diabetes mellitus and chronic kidney disease and might contribute to disease by stimulating inflammation. Although urate has been classically viewed as an antioxidant with beneficial effects, new data suggest that both crystalline and soluble urate activate various pro-inflammatory pathways. This Review summarizes what is known about the role of urate in the inflammatory response. Further research is needed to define the role of asymptomatic hyperuricaemia in these pro-inflammatory pathways.
Collapse
Affiliation(s)
- Leo A B Joosten
- Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
- Department of Internal Medicine and Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Tania O Crişan
- Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Petter Bjornstad
- Department of Medicine of the University of Colorado School of Medicine of the University Hospital, Aurora, CO, USA
| | - Richard J Johnson
- Department of Medicine of the University of Colorado School of Medicine of the University Hospital, Aurora, CO, USA.
| |
Collapse
|
22
|
Dreh, a long noncoding RNA repressed by metformin, regulates glucose transport in C2C12 skeletal muscle cells. Life Sci 2019; 236:116906. [PMID: 31614147 DOI: 10.1016/j.lfs.2019.116906] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 09/13/2019] [Accepted: 09/23/2019] [Indexed: 12/20/2022]
Abstract
AIMS The anti-hyperglycemic action of metformin on skeletal muscles is presently unclear. Long noncoding RNAs (lncRNAs) are implicated in multiple cellular functions. This study aims to explore the role of lncRNAs in the glucometabolic action of metformin on skeletal muscle cells. MAIN METHODS Metformin accumulation was assessed using [14C]-metformin. A lncRNA array was used to investigate metformin-regulated lncRNAs in C2C12 skeletal muscle cells. Knockdown studies were applied to evaluate the function of lncRNA Dreh. A colorimetric assay was used for the measurement of medium glucose concentration; glucose transport was assessed using [3H]-2-deoxyglucose; real-time PCR was used for RNA expression analysis, and western blotting was used to assess protein expression in myotubes. A Dreh overexpression plasmid was transfected into the cells. KEY FINDINGS Metformin accumulated in C2C12 myotubes. Metformin reduced medium glucose concentration and repressed lncRNA Dreh expression in the myotubes. Knockdown of Dreh in the myotubes resulted in reduced glucose concentration in the culture medium, increased glucose transport, and increased levels of GLUT4 protein in the plasma membrane. Overexpression of Dreh attenuated the glucose-lowering effect of metformin in myotubes. SIGNIFICANCE The glucoregulatory actions of metformin are mediated in part by a lncRNA, Dreh, in the skeletal muscle cells. Dreh is a novel regulator for glucose transport and could be a therapeutic target for diabetes.
Collapse
|
23
|
Xiao J, Zhu S, Guan H, Zheng Y, Li F, Zhang X, Guo H, Wang X, Ye Z. AMPK alleviates high uric acid-induced Na +-K +-ATPase signaling impairment and cell injury in renal tubules. Exp Mol Med 2019; 51:1-14. [PMID: 31118410 PMCID: PMC6531502 DOI: 10.1038/s12276-019-0254-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 01/30/2019] [Accepted: 02/08/2019] [Indexed: 12/22/2022] Open
Abstract
One of the mechanisms in hyperuricemia (HUA)-induced renal tubular injury is the impairment of Na+-K+-ATPase (NKA) signaling, which further triggers inflammation, autophagy, and mitochondrial dysfunction and leads to cell injury. Here, we used RNA sequencing to screen the most likely regulators of NKA signaling and found that the liver kinase B1(LKB1)/adenosine monophosphate (AMP)-activated protein kinase (AMPK)/ mammalian target of rapamycin (mTOR) pathway was the most abundantly enriched pathway in HUA. AMPK is a key regulator of cell energy metabolism; hence, we examined the effect of AMPK on HUA-induced dysregulation of NKA signaling and cell injury. We first detected AMPK activation in high uric acid (UA)-stimulated proximal tubular epithelial cells (PTECs). We further found that sustained treatment with the AMPK activator 5-aminoimidazole-4-carboxamide 1-β-d-ribofuranoside (AICAR), but not the AMPK inhibitor Compound C, significantly alleviated UA-induced reductions in NKA activity and NKA α1 subunit expression on the cell membrane by reducing NKA degradation in lysosomes; sustained AICAR treatment also significantly alleviated activation of the NKA downstream molecules Src and interleukin-1β (IL-1β) in PTECs. AICAR further alleviated high UA-induced apoptosis, autophagy, and mitochondrial dysfunction. Although AMPK activation by metformin did not reduce serum UA levels in hyperuricemic rats, it significantly alleviated HUA-induced renal tubular injury and NKA signaling impairment in vivo with effects similar to those of febuxostat. Our study suggests that AMPK activation may temporarily compensate for HUA-induced renal injury. Sustained AMPK activation could reduce lysosomal NKA degradation and maintain NKA function, thus alleviating NKA downstream inflammation and protecting tubular cells from high UA-induced renal tubular injury. High serum levels of uric acid cause kidney tissue damage through cellular processes that have now been identified by researchers in China. Uric acid is a common component of urine, but causes damage if it is present in high levels in the blood (hyperuricemia). While investigating the mechanisms behind hyperuricemia, Zhibin Ye and co-workers at Fudan University in Shanghai recently showed that impairment of the Na+-K+-ATPase (NKA) signaling pathway, which regulates uric acid transportation through the kidneys, is a crucial feature of renal damage progression. The team have now shown that NKA is regulated by the AMP-activated protein kinase (AMPK) pathway, and that AMPK is enriched during the initial phases of hyperuricemia. Studies on rat models indicated that sustained AMPK activation restored NKA signaling, limiting damage from hyperuricemia.
Collapse
Affiliation(s)
- Jing Xiao
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, People's Republic of China
| | - Sibo Zhu
- School of Life Sciences, Fudan University, Shanghai, People's Republic of China
| | - Haochen Guan
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, People's Republic of China
| | - Yuqi Zheng
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, People's Republic of China
| | - Fengqin Li
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, People's Republic of China
| | - Xiaoli Zhang
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, People's Republic of China
| | - Hui Guo
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, People's Republic of China
| | - Xiaojun Wang
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, People's Republic of China
| | - Zhibin Ye
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, Shanghai, People's Republic of China. .,Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
24
|
A long noncoding RNA LOC103690121 promotes hippocampus neuronal apoptosis in streptozotocin-induced type 1 diabetes. Neurosci Lett 2019; 703:11-18. [PMID: 30851305 DOI: 10.1016/j.neulet.2019.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/07/2019] [Accepted: 03/05/2019] [Indexed: 12/16/2022]
Abstract
Diabetes related cognitive impairment is a severe complication. The diabetes-induced cognitive impairment is associated with insulin resistance and glucose-induced neuron apoptosis in the brain. We intended to investigate the association of long non-coding RNAs with diabetes-induced cognitive impairment in rats. Here, Type 1diabetes (T1D) rat model was induced using streptozotocin (STZ). The diabetic rats showed significant cognitive dysfunction, with increased latency period to find the hidden platform during morris water maze test. The brain injury and reduced neuronsin STZ-induced diabetic rats was determined using hematoxylin and eosin staining and Nissl's staining. We performed the LncRNA microarray analysis and identified 101 differentially expressed lncRNAs in streptozotocin (STZ)-induced type 1 diabetes (T1D) comparing with control. Among these lncRNA, LOC103690121 was upregulated. in vitro glucose treatment in hippocampal neurons showed LOC103690121 and neuron apoptosis was increased by glucose treatment. Transfection experiments showed LOC103690121 overexpression promoted neuron apoptosis, and its inhibition suppressed glucose-induced apoptosis. Western blot analysis showed that the expression profiles of apoptosis related proteins (cleaved-caspase-3, -8, -9, and Bax) were in line with LOC103690121 expression, while the profiles of Bcl-2 and PI3K/Akt signaling pathway was contrast to LOC103690121 expression. In conclusion, the results of our study confirmed lncRNA LOC103690121 promoted STZ-induced cognitive impairment in diabetic rats by promoting neuron apoptosis through PI3K/Akt signaling pathway.
Collapse
|
25
|
Allopurinol potentiates the hepatoprotective effect of metformin and vitamin E in fructose-induced fatty liver in rats. Clin Exp Hepatol 2019; 5:65-74. [PMID: 30915409 PMCID: PMC6431087 DOI: 10.5114/ceh.2019.83159] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/14/2018] [Indexed: 02/07/2023] Open
Abstract
Aim of the study Non-alcoholic fatty liver disease (NAFLD) is a challenging health problem. Hyperuricemia is a key player in the pathogenesis of NAFLD. This study investigated the effect of allopurinol (uric acid synthesis inhibitor) in combination with metformin and vitamin E in prevention of fructose induced-fatty liver in rats. Material and methods Rats were divided into 7 groups: control group, fructose group (model group of NAFLD), allopurinol-treated group, metformin-treated group, vitamin E-treated group, metformin plus vitamin E-treated group and a combination group (received allopurinol plus metformin plus vitamin E). Development of NAFLD was assessed biochemically by serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) as well as by histopathological examination. Oxidative stress parameters [reduced glutathione (GSH), superoxide dismutase (SOD), malondialdehyde (MDA)], and the inflammatory mediators tumor necrosis factor α (TNF-α) and inducible nitric oxide synthase (iNOS) were assessed, along with serum levels of uric acid and triglyceride (TG). Results Combination of allopurinol plus metformin plus vitamin E significantly attenuated fatty changes compared to their respective monotherapy. Interestingly, though all treated groups showed significant attenuation in the oxidative stress markers, TNF-α level and iNOS immunostaining in hepatic tissue, along with a significant decrease in the levels of uric acid and TG, the combination group showed a further significant decrease in the serum level of uric acid and iNOS immunostaining compared to other treated regimens. Conclusions Allopurinol synergistically increases the protective effect of metformin and vitamin E in treatment of NAFLD, namely via reduction of uric acid synthesis and iNOS expression.
Collapse
|
26
|
Brunetti A, Arcidiacono B, Foti DP, Semple RK. Editorial: Transcriptional Regulation of Glucose Metabolism: Gaps and Controversies. Front Endocrinol (Lausanne) 2019; 10:629. [PMID: 31620085 PMCID: PMC6759599 DOI: 10.3389/fendo.2019.00629] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 08/30/2019] [Indexed: 12/26/2022] Open
Affiliation(s)
- Antonio Brunetti
- Department of Health Sciences, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
- *Correspondence: Antonio Brunetti
| | - Biagio Arcidiacono
- Department of Health Sciences, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Daniela Patrizia Foti
- Department of Health Sciences, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Robert K. Semple
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
- Robert K. Semple
| |
Collapse
|
27
|
Xu D, Wang L, Jiang Z, Zhao G, Hassan HM, Sun L, Fan S, Zhou Z, Zhang L, Wang T. A new hypoglycemic mechanism of catalpol revealed by enhancing MyoD/MyoG-mediated myogenesis. Life Sci 2018; 209:313-323. [PMID: 30118770 DOI: 10.1016/j.lfs.2018.08.028] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 07/26/2018] [Accepted: 08/11/2018] [Indexed: 01/08/2023]
Abstract
AIMS Enhancing myogenesis has been identified as a possible target to improve insulin sensitivity and protect against metabolic diseases. Catalpol, an iridoid glycoside, has been shown to exert a hypoglycaemic effect by improvement of insulin sensitivity; however, the underlying mechanism remains unknown. In this study, we tested whether catalpol has the potential to improve insulin sensitivity by augmenting myogenesis. MAIN METHODS We examined the hypoglycaemic mechanism of catalpol in db/db mice and C2C12 cells. db/db mice were treated with catalpol (200 mg/kg) for 8 consecutive weeks. Serum analysis, skeletal muscle performance and histology, and gene and protein expression were performed. In vitro glucose uptake, gene and protein expression were determined, and small interfering RNA was used to identify the underlying hypoglycaemic mechanism of catalpol. KEY FINDINGS In this study, we tested whether catalpol has the potential to improve skeletal insulin sensitivity by augmenting myogenesis, in which we found that, catalpol treatment in db/db mice lowered blood glucose and improved insulin sensitivity via activation of phosphatidylinositol‑3‑Kinase (PI3K)/protein kinase B (AKT) pathway. Moreover, catalpol-treated mice exhibited enhanced myogenesis, as evidenced by increased myogenic differentiation (MyoD), myogenin (MyoG) and myosin heavy chain (MHC) expressions. The in vitro experimental results showed that both catalpol and metformin enhanced glucose uptake via activation of PI3K/AKT pathway. However, unlike metformin, the PI3K/AKT pathway activation by catalpol was dependent on enhanced MyoD/MyoG-mediated myogenesis. SIGNIFICANCE Improvement of insulin sensitivity by enhancing MyoD/MyoG-mediated myogenesis may constitute a new therapeutic approach for treating type 2 diabetes.
Collapse
Affiliation(s)
- Dengqiu Xu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Lu Wang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Zhenzhou Jiang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing 210009, China
| | - Guolin Zhao
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Hozeifa M Hassan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Lixin Sun
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Sisi Fan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Zhixing Zhou
- Tianjin Key Laboratory of Molecular Design and Drug Discovery, Tianjin Institute of Pharmaceutical Research, Tianjin 300193, China
| | - Luyong Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Center for Drug Screening and Pharmacodynamics Evaluation, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing 210009, China.
| | - Tao Wang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
28
|
Metabotypes of response to bariatric surgery independent of the magnitude of weight loss. PLoS One 2018; 13:e0198214. [PMID: 29856816 PMCID: PMC5983508 DOI: 10.1371/journal.pone.0198214] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 05/15/2018] [Indexed: 12/18/2022] Open
Abstract
Objective Bariatric surgery is considered the most efficient treatment for morbid obesity and its related diseases. However, its role as a metabolic modifier is not well understood. We aimed to determine biosignatures of response to bariatric surgery and elucidate short-term metabolic adaptations. Methods We used a LC- and FIA-ESI-MS/MS approach to quantify acylcarnitines, (lyso)phosphatidylcholines, sphingomyelins, amino acids, biogenic amines and hexoses in serum samples of subjects with morbid obesity (n = 39) before and 1, 3 and 6 months after bariatric surgery. K-means cluster analysis allowed to distinguish metabotypes of response to bariatric surgery. Results For the first time, global metabolic changes following bariatric surgery independent of the baseline health status of the subjects have been revealed. We identify two metabolic phenotypes (metabotypes) at the interval 6 months-baseline after surgery, which presented differences in the levels of compounds of urea metabolism, gluconeogenic precursors and (lyso)phospholipid particles. Clinically, metabotypes were different in terms of the degree of improvement in insulin resistance, cholesterol, low-density lipoproteins and uric acid independent of the magnitude of weight loss. Conclusions This study opens new perspectives and new hypotheses on the metabolic benefits of bariatric surgery and understanding of the biology of obesity and its associated diseases.
Collapse
|
29
|
How AMPK and PKA Interplay to Regulate Mitochondrial Function and Survival in Models of Ischemia and Diabetes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4353510. [PMID: 29391924 PMCID: PMC5748092 DOI: 10.1155/2017/4353510] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 11/02/2017] [Indexed: 12/17/2022]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) is a conserved, redox-activated master regulator of cell metabolism. In the presence of oxidative stress, AMPK promotes cytoprotection by enhancing the conservation of energy by suppressing protein translation and by stimulating autophagy. AMPK interplays with protein kinase A (PKA) to regulate oxidative stress, mitochondrial function, and cell survival. AMPK and dual-specificity A-kinase anchoring protein 1 (D-AKAP1), a mitochondrial-directed scaffold of PKA, interact to regulate mitochondrial function and oxidative stress in cardiac and endothelial cells. Ischemia and diabetes, a chronic disease that increases the onset of cardiovascular diseases, suppress the cardioprotective effects of AMPK and PKA. Here, we review the molecular mechanisms by which AMPK and D-AKAP1/PKA interplay to regulate mitochondrial function, oxidative stress, and signaling pathways that prime endothelial cells, cardiac cells, and neurons for cytoprotection against oxidative stress. We discuss recent literature showing how temporal dynamics and localization of activated AMPK and PKA holoenzymes play a crucial role in governing cellular bioenergetics and cell survival in models of ischemia, cardiovascular diseases, and diabetes. Finally, we propose therapeutic strategies that tout localized PKA and AMPK signaling to reverse mitochondrial dysfunction, oxidative stress, and death of neurons and cardiac and endothelial cells during ischemia and diabetes.
Collapse
|
30
|
Leng J, Wang L, Wang J, Li W, Liu H, Zhang S, Li L, Tian H, Xun P, Yang X, Yu Z, Hu G. Uric acid and diabetes risk among Chinese women with a history of gestational diabetes mellitus. Diabetes Res Clin Pract 2017; 134:72-79. [PMID: 28993154 PMCID: PMC5723553 DOI: 10.1016/j.diabres.2017.09.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 09/02/2017] [Accepted: 09/27/2017] [Indexed: 01/06/2023]
Abstract
AIMS To assess the association of uric acid (UA) with the risks of postpartum type 2 diabetes and prediabetes among women with prior gestational diabetes mellitus (GDM). METHODS We performed a cross-sectional study of 1262 GDM women at 1-5 years after delivery using the baseline data from the Tianjin Gestational Diabetes Mellitus Prevention Program. Logistic regression models were used to estimate the association of different levels of serum UA with the risks of type 2 diabetes and prediabetes. RESULTS The multivariable-adjusted odds ratios (ORs) across quartiles of serum UA were 1.00, 1.23 (95% confidence interval [CI] 0.55-2.78), 2.05 (95% CI 0.96-4.39), and 3.17 (95% CI 1.54-6.55) (Ptrend < 0.001) for type 2 diabetes, and 1.00, 1.50 (95% CI 1.03-2.19), 2.28 (95% CI 1.58-3.30), and 2.88 (95% CI 1.99-4.17) (Ptrend < 0.001) for prediabetes, respectively. Restricted cubic splines models showed positive linear associations of serum UA as a continuous variable with the risks of type 2 diabetes and prediabetes. This positive association was significant when stratified by healthy weight and overweight participants. CONCLUSIONS Serum UA levels have a graded positive association with the risks of type 2 diabetes and prediabetes among Chinese with a history of GDM.
Collapse
Affiliation(s)
- Junhong Leng
- Tianjin Women's and Children's Health Center, Tianjin, China
| | - Leishen Wang
- Tianjin Women's and Children's Health Center, Tianjin, China
| | - Jing Wang
- Tianjin Women's and Children's Health Center, Tianjin, China; Chronic Disease Epidemiology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Weiqin Li
- Tianjin Women's and Children's Health Center, Tianjin, China
| | - Huikun Liu
- Tianjin Women's and Children's Health Center, Tianjin, China
| | - Shuang Zhang
- Tianjin Women's and Children's Health Center, Tianjin, China
| | - Lili Li
- Tianjin Women's and Children's Health Center, Tianjin, China
| | - Huiguang Tian
- Tianjin Women's and Children's Health Center, Tianjin, China
| | - Pengcheng Xun
- Department of Epidemiology and Biostatistics, School of Public Health-Bloomington, Indiana University, Bloomington, IN, USA
| | - Xilin Yang
- Department of Epidemiology and Biostatistics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin, China
| | - Zhijie Yu
- Population Cancer Research Program, Dalhousie University, 1494 Carlton Street, Halifax, NS, Canada
| | - Gang Hu
- Chronic Disease Epidemiology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA.
| |
Collapse
|
31
|
Wang H, Zhu C, Ying Y, Luo L, Huang D, Luo Z. Metformin and berberine, two versatile drugs in treatment of common metabolic diseases. Oncotarget 2017. [PMID: 29515798 PMCID: PMC5839379 DOI: 10.18632/oncotarget.20807] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Metformin has been used as a glucose lowering drug for several centuries and is now a first-line drug for type 2 diabetes mellitus (T2DM). Since the discovery that it activates AMP-activated protein kinase (AMPK) and reduces risk of cancer, metformin has drawn great attentions. Another drug, berberine, extracted from berberis vulgaris L. (root), was an ancient herbal medicine in treating diarrhea. Ongoing experimental and clinical studies have illuminated great potential of berberine in regulation of glucose and lipid homeostasis, cancer growth and inflammation. Furthermore, the lipid lowering effect of berberine is comparable to those conventional lipid drugs but with low toxicity. Therefore, it is right time to transform beneficial effects of berberine into therapeutic practice. Metformin and berberine share many features in actions despite different structure and both could be excellent drugs in treating T2DM, obesity, cardiac diseases, tumour, as well as inflammation. Since these disorders are often connected and comprise common pathogenic factors that could be targeted by the two drugs, understanding their actions can give us rationale for expansion of their clinical uses.
Collapse
Affiliation(s)
- Haoran Wang
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Hospital of Nanchang University, Nanchang, China
| | - Chen Zhu
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Hospital of Nanchang University, Nanchang, China
| | - Ying Ying
- Jiangxi Provincial Key Laboratory of Tumour Pathogenesis and Molecular Pathology, Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang, China
| | - Lingyu Luo
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Hospital of Nanchang University, Nanchang, China
| | - Deqiang Huang
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Hospital of Nanchang University, Nanchang, China
| | - Zhijun Luo
- Jiangxi Provincial Key Laboratory of Tumour Pathogenesis and Molecular Pathology, Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang, China.,Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
32
|
Wang J, Gao Y, Duan L, Wei S, Liu J, Tian L, Quan J, Zhang Q, Liu J, Yang J. Metformin ameliorates skeletal muscle insulin resistance by inhibiting miR-21 expression in a high-fat dietary rat model. Oncotarget 2017; 8:98029-98039. [PMID: 29228671 PMCID: PMC5716711 DOI: 10.18632/oncotarget.20442] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 07/25/2017] [Indexed: 12/19/2022] Open
Abstract
Insulin resistance (IR) plays a major role in the pathogenesis of abdominal obesity, hypertension, coronary heart disease, atherosclerosis and diabetes. miR-21 and TGF-β/smads is closely related to IR. However, it remained elusive whether metformin improved skeletal muscle insulin resistance (IRSM) by regulating miR-21 and its target signal TGF-β1/smads expression. In this study, high-fat diet rats with IR model and IR-skeletal muscle L6 cells (L6-SMCs) model were established, insulin sensitive index (ISI) and Homeostasis model assessment of IR (HOMA-IR) were applied, miR-21 and TGF-β1/smads mRNA expression were examined by RT-PCR, smad3 and smad7 protein were detected by western-blotting and laser scanning confocal microscopy (LSCM), the valid target of miR-21 was detected by luciferase reporter gene assay. Here, we found that metformin dose-dependently decreased miR-21 expression, accompanied by the decrease of HOMA-IR and the increase of HOMA-ISI. Luciferase report gene assay showed that smad7 was an effective target of miR-21. miR-21 overexpression directly downregulated smad7 and indirectly upregulated smad3 expression. Interestingly, miR-21 expression positively correlated with HOMA-IR and negatively correlated with HOMA-ISI. In conclusion, our results demonstrated that metformin improved IRSM by inhibiting miR-21 expression, and that miR-21 may be one of the therapeutic targets for IR.
Collapse
Affiliation(s)
- Jinyang Wang
- Department of Endocrinology, Gansu Provincial People's hospital, Lanzhou, China.,Gansu Provincial Key Laboratory of Endocrine and metabolism, Lanzhou, China
| | - Yanbin Gao
- School of Traditional Chinese medical, Capital Medical University, Beijing, China.,Beijing Key Laboratory of TCM Collateral Disease Theory Research, Beijing, China
| | - Lijun Duan
- Department of Gynecology and Obstetrics, Gansu Provincial People's Hospital, Lanzhou, China
| | - Suhong Wei
- Department of Endocrinology, Gansu Provincial People's hospital, Lanzhou, China.,Gansu Provincial Key Laboratory of Endocrine and metabolism, Lanzhou, China
| | - Jing Liu
- Department of Endocrinology, Gansu Provincial People's hospital, Lanzhou, China.,Gansu Provincial Key Laboratory of Endocrine and metabolism, Lanzhou, China
| | - Liming Tian
- Department of Endocrinology, Gansu Provincial People's hospital, Lanzhou, China.,Gansu Provincial Key Laboratory of Endocrine and metabolism, Lanzhou, China
| | - Jinxing Quan
- Department of Endocrinology, Gansu Provincial People's hospital, Lanzhou, China.,Gansu Provincial Key Laboratory of Endocrine and metabolism, Lanzhou, China
| | - Qi Zhang
- Department of Endocrinology, Gansu Provincial People's hospital, Lanzhou, China.,Gansu Provincial Key Laboratory of Endocrine and metabolism, Lanzhou, China
| | - Juxiang Liu
- Department of Endocrinology, Gansu Provincial People's hospital, Lanzhou, China.,Gansu Provincial Key Laboratory of Endocrine and metabolism, Lanzhou, China
| | - Jinkui Yang
- Department of Endocrinology, Beijing Tongren hospital of Capital Medical University, Beijing, China
| |
Collapse
|