1
|
Asthana P, Wong HLX. Preventing obesity, insulin resistance and type 2 diabetes by targeting MT1-MMP. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167081. [PMID: 38367902 DOI: 10.1016/j.bbadis.2024.167081] [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: 12/28/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/19/2024]
Abstract
Obesity is one of the predominant risk factors for type 2 diabetes. Despite all the modern advances in medicine, an effective drug treatment for obesity without overt side effects has not yet been found. The discovery of growth and differentiation factor 15 (GDF15), an appetite-regulating hormone, created hopes for the treatment of obesity. However, an insufficient understanding of the physiological regulation of GDF15 has been a major obstacle to mitigating GDF15-centric treatment of obesity. Our recent studies revealed how a series of proteolytic events predominantly mediated by membrane-type 1 matrix metalloproteinase (MT1-MMP/MMP14), a key cell-surface metalloproteinase involved in extracellular remodeling, contribute to the pathogenesis of metabolic disorders, including obesity and diabetes. The MT1-MMP-mediated cleavage of the GDNF family receptor-α-like (GFRAL), a key neuronal receptor of GDF15, controls the satiety center in the hindbrain, thereby regulating non-homeostatic appetite and bodyweight changes. Furthermore, increased activation of MT1-MMP does not only lead to increased risk of obesity, but also causes age-associated insulin resistance by cleaving Insulin Receptor in major metabolic tissues. Importantly, inhibition of MT1-MMP effectively protects against obesity and diabetes, revealing the therapeutic potential of targeting MT1-MMP for the management of metabolic disorders.
Collapse
Affiliation(s)
- Pallavi Asthana
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong
| | | |
Collapse
|
2
|
Qin Y, Chen J, Qian D, Li Z, Zhang L, Ma Q. Excessive Tryptophan and Phenylalanine Induced Pancreatic Injury and Glycometabolism Disorder in Grower-finisher Pigs. J Nutr 2024; 154:1333-1346. [PMID: 38582698 DOI: 10.1016/j.tjnut.2024.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/03/2024] [Accepted: 01/17/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND The increase in circulating insulin levels is associated with the onset of type 2 diabetes (T2D), and the levels of branched-chain amino acids and aromatic amino acids (AAAs) are altered in T2D, but whether AAAs play a role in insulin secretion and signaling remains unclear. OBJECTIVES This study aimed to investigate the effects of different AAAs on pancreatic function and on the use of insulin in finishing pigs. METHODS A total of 18 healthy finishing pigs (Large White) with average body weight of 100 ± 1.15 kg were randomly allocated to 3 dietary treatments: Con, a normal diet supplemented with 0.68% alanine; Phe, a normal diet supplemented with 1.26% phenylalanine; and Trp, a normal diet supplemented with 0.78% tryptophan. The 3 diets were isonitrogenous. There were 6 replicates in each group. RESULTS Herein, we investigated the effects of tryptophan and phenylalanine on pancreatic function and the use of insulin in finishing pigs and found that the addition of tryptophan and phenylalanine aggravated pancreatic fat deposition, increased the relative content of saturated fatty acids, especially palmitate (C16:0) and stearate (C18:0), and the resulting lipid toxicity disrupted pancreatic secretory function. We also found that tryptophan and phenylalanine inhibited the growth and secretion of β-cells, downregulated the gene expression of the PI3K/Akt pathway in the pancreas and liver, and reduced glucose utilization in the liver. CONCLUSIONS Using fattening pigs as a model, multiorgan combined analysis of the insulin-secreting organ pancreas and the main insulin-acting organ liver, excessive intake of tryptophan and phenylalanine will aggravate pancreatic damage leading to glucose metabolism disorders, providing new evidence for the occurrence and development of T2D.
Collapse
Affiliation(s)
- Yingjie Qin
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Jiayi Chen
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Dali Qian
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Zhongyu Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Licong Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Qingquan Ma
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China.
| |
Collapse
|
3
|
Greeny A, Nair A, Sadanandan P, Satarker S, Famurewa AC, Nampoothiri M. Epigenetic Alterations in Alzheimer's Disease: Impact on Insulin Signaling and Advanced Drug Delivery Systems. BIOLOGY 2024; 13:157. [PMID: 38534427 DOI: 10.3390/biology13030157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/25/2024] [Accepted: 02/26/2024] [Indexed: 03/28/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition that predominantly affects the hippocampus and the entorhinal complex, leading to memory lapse and cognitive impairment. This can have a negative impact on an individual's behavior, speech, and ability to navigate their surroundings. AD is one of the principal causes of dementia. One of the most accepted theories in AD, the amyloid β (Aβ) hypothesis, assumes that the buildup of the peptide Aβ is the root cause of AD. Impaired insulin signaling in the periphery and central nervous system has been considered to have an effect on the pathophysiology of AD. Further, researchers have shifted their focus to epigenetic mechanisms that are responsible for dysregulating major biochemical pathways and intracellular signaling processes responsible for directly or indirectly causing AD. The prime epigenetic mechanisms encompass DNA methylation, histone modifications, and non-coding RNA, and are majorly responsible for impairing insulin signaling both centrally and peripherally, thus leading to AD. In this review, we provide insights into the major epigenetic mechanisms involved in causing AD, such as DNA methylation and histone deacetylation. We decipher how the mechanisms alter peripheral insulin signaling and brain insulin signaling, leading to AD pathophysiology. In addition, this review also discusses the need for newer drug delivery systems for the targeted delivery of epigenetic drugs and explores targeted drug delivery systems such as nanoparticles, vesicular systems, networks, and other nano formulations in AD. Further, this review also sheds light on the future approaches used for epigenetic drug delivery.
Collapse
Affiliation(s)
- Alosh Greeny
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Ayushi Nair
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Amrita Health Science Campus, Kochi 682041, India
| | - Prashant Sadanandan
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Amrita Health Science Campus, Kochi 682041, India
| | - Sairaj Satarker
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medical Sciences, Alex Ekwueme Federal University, Ndufu-Alike, Ikwo 482123, Nigeria
| | - Madhavan Nampoothiri
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| |
Collapse
|
4
|
Wang S, Wu J, Chen Z, Wu W, Lu L, Cheng Y, Li S, Chen L, Tan X, Yang L, Wang C, Song Y. DNA methylation reprogramming mediates transgenerational diabetogenic effect induced by early-life p,p'-DDE exposure. CHEMOSPHERE 2024; 349:140907. [PMID: 38092165 DOI: 10.1016/j.chemosphere.2023.140907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/18/2023] [Accepted: 12/03/2023] [Indexed: 12/19/2023]
Abstract
Increasing evidence shows that an adverse environment during the early fetal development can affect the epigenetic modifications on a wide range of diabetes-related genes, leading to an increased diabetic susceptibility in adulthood or even in subsequent generations. p,p'-Dichlorodiphenoxydichloroethylene (p,p'-DDE) is a break-down product of the pesticide dichlorodiphenyltrichloroethane (DDT). p,p'-DDE has been associated with various health concerns, such as diabetogenic effect. However, the precise molecular mechanism remains unclear. In this study, p,p'-DDE was given by gavage to pregnant rat dams from gestational day (GD) 8 to GD15 to generate male germline to investiagate the transgenerational effects. We found that early-life p,p'-DDE exposure increased the transgenerational diabetic susceptibility through male germline inheritance. In utero exposure to p,p'-DDE altered the sperm DNA methylome in F1 progeny, and a significant number of those differentially methylated genes could be inherited by F2 progeny. Furthermore, early-life p,p'-DDE exposure altered DNA methylation in glucose metabolic genes Gck and G6pc in sperm and the methylation modification were also found in liver of the next generation. Our study demonstrate that DNA methylation plays a critical role in mediating transgenerational diabetogenic effect induced by early-life p,p'-DDE exposure.
Collapse
Affiliation(s)
- Shuo Wang
- School of Public Health, Hangzhou Normal University, 2318 Yuhangtang Road, Hangzhou, China
| | - Jingjing Wu
- School of Public Health, Hangzhou Normal University, 2318 Yuhangtang Road, Hangzhou, China
| | - Zhong Chen
- Center for Genomics, Loma Linda University School of Medicine, 11021 Campus Street, Loma Linda, CA, 92350, USA
| | - Wei Wu
- School of Public Health, Hangzhou Normal University, 2318 Yuhangtang Road, Hangzhou, China
| | - Liping Lu
- School of Public Health, Hangzhou Normal University, 2318 Yuhangtang Road, Hangzhou, China
| | - Yuzhou Cheng
- School of Public Health, Hangzhou Normal University, 2318 Yuhangtang Road, Hangzhou, China
| | - Shuqi Li
- School of Public Health, Hangzhou Normal University, 2318 Yuhangtang Road, Hangzhou, China
| | - Liangjing Chen
- School of Public Health, Hangzhou Normal University, 2318 Yuhangtang Road, Hangzhou, China
| | - Xiaohua Tan
- School of Public Health, Hangzhou Normal University, 2318 Yuhangtang Road, Hangzhou, China
| | - Lei Yang
- School of Public Health, Hangzhou Normal University, 2318 Yuhangtang Road, Hangzhou, China
| | - Charles Wang
- Center for Genomics, Loma Linda University School of Medicine, 11021 Campus Street, Loma Linda, CA, 92350, USA
| | - Yang Song
- School of Public Health, Hangzhou Normal University, 2318 Yuhangtang Road, Hangzhou, China.
| |
Collapse
|
5
|
Feng L, Chen C, Guo Q, Chen L, Yang W. Improvement of early-phase insulin secretion is an independent factor for achieving glycaemic control: A pooled analysis of SEED and DAWN study. Diabetes Obes Metab 2024; 26:745-753. [PMID: 37985364 DOI: 10.1111/dom.15370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/25/2023] [Accepted: 11/01/2023] [Indexed: 11/22/2023]
Abstract
AIM To investigate the effect of improving early phase insulin secretion function for glycaemic control in patients with type 2 diabetes mellitus treated with a new class of antidiabetic drug dorzagliatin. MATERIALS AND METHODS Early insulin secretion function was studied in 726 participants of which 414 were treated with dorzagliatin in the SEED and DAWN study. The early insulinogenic index (IGI30min ) and disposition index (DI) were used to assess early-phase insulin secretion function in this study. Logistic regression analysis was performed to verify the importance of IGI30min and DI indices for achieving effective glycaemic control. RESULTS The reduction in HbA1c has a significant correlation with the improvement of IGI30min for patients that received 24 weeks of dorzagliatin treatment (p < .001), and this correlation was not observed in the placebo group (p = .364). In the dorzagliatin treatment group, the responders showed significant improvements in homeostasis model assessment 2-β, IGI30min and DI compared with the non-responders. Logistic regression analysis revealed that the odds ratio (OR) for achieving glycaemic control was 1.28 (95% CI 1.14-1.43) for baseline IGI30min , and 1.24 (95% CI 1.14-1.35) for the 24-week incremental IGI30min from baseline. The OR for baseline DI and 24-week changes in DI from baseline were 1.39 (95% CI 1.2-1.6) and 1.30 (95% CI 1.19-1.43) respectively. The timing of insulin secretion analysis showed the significant contribution of early-phase insulin secretion, rather than late-phase insulin secretion, to postprandial glucose control with the OR for the incremental IGI30min and IGI2h to postprandial glucose control were 1.3 (95% CI 1.19-1.42) and 1 (95% CI 1-1.01) respectively. CONCLUSIONS Restoring the impaired early-phase insulin secretion function in patients with type 2 diabetes mellitus is a critical factor for improving the glycaemic control by dorzagliatin treatment.
Collapse
Affiliation(s)
| | | | | | - Li Chen
- Hua Medicine, Shanghai, China
| | - Wenying Yang
- Japan-China Friendship Hospital, Beijing, China
- Taikang Yanyuan Rehabilitation Hospital, Beijing, China
| |
Collapse
|
6
|
Chen L, Zhang J, Sun Y, Zhao Y, Liu X, Fang Z, Feng L, He B, Zou Q, Tracey GJ. A phase I open-label clinical trial to study drug-drug interactions of Dorzagliatin and Sitagliptin in patients with type 2 diabetes and obesity. Nat Commun 2023; 14:1405. [PMID: 36918550 PMCID: PMC10014962 DOI: 10.1038/s41467-023-36946-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 02/22/2023] [Indexed: 03/16/2023] Open
Abstract
This is a phase 1, open-label, single-sequence, multiple-dose, single-center trial conducted in the US (NCT03790839), to evaluate the clinical pharmacokinetics, safety and pharmacodynamics of dorzagliatin co-administered with sitagliptin in patients with T2D and obesity. The trial has completed. 15 patients with T2D and obesity were recruited and treated with sitagliptin 100 mg QD on Day 1-5, followed by a combination of sitagliptin 100 mg QD with dorzagliatin 75 mg BID at second stage on Day 6-10 and the third stage of dorzagliatin 75 mg BID alone on Day 11-15. Primary outcomes include pharmacokinetic geometric mean ratio (GMR), safety and tolerability. Secondary outcomes include the incremental area under the curve for 4 hours post oral glucose tolerance test (iAUC) of pharmacodynamic biomarkers and glucose sensitivity. GMR for AUC0-24h and Cmax were 92.63 (90% CI, 85.61, 100.22) and 98.14 (90% CI, 83.73, 115.03) in combination/sitagliptin, and 100.34 (90% CI, 96.08, 104.79) and 102.34 (90% CI, 86.92, 120.50) in combination/dorzagliatin, respectively. Combination treatment did not increase the adverse events and well-tolerated in T2D patients. Lack of clinically meaningful pharmacokinetic interactions between dorzagliatin and sitagliptin, and an improvement of glycemic control under combination potentially support their co-administration for diabetes management.
Collapse
Affiliation(s)
- Li Chen
- Hua Medicine (Shanghai) Limited, Shanghai, China.
| | - Jiayi Zhang
- Hua Medicine (Shanghai) Limited, Shanghai, China
| | - Yu Sun
- Hua Medicine (Shanghai) Limited, Shanghai, China
| | - Yu Zhao
- Hua Medicine (Shanghai) Limited, Shanghai, China
| | - Xiang Liu
- Hua Medicine (Shanghai) Limited, Shanghai, China
| | - Zhiyin Fang
- Hua Medicine (Shanghai) Limited, Shanghai, China
| | - Lingge Feng
- Hua Medicine (Shanghai) Limited, Shanghai, China
| | - Bin He
- Hua Medicine (Shanghai) Limited, Shanghai, China
| | - Quanfei Zou
- Hua Medicine (Shanghai) Limited, Shanghai, China
| | | |
Collapse
|
7
|
Jiang C, Hu Y, Wang S, Chen C. Emerging trends in DNA and RNA methylation modifications in type 2 diabetes mellitus: a bibliometric and visual analysis from 1992 to 2022. Front Endocrinol (Lausanne) 2023; 14:1145067. [PMID: 37201099 PMCID: PMC10187586 DOI: 10.3389/fendo.2023.1145067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/30/2023] [Indexed: 05/20/2023] Open
Abstract
Background Type 2 diabetes mellitus (T2DM) is a pathological metabolic disorder induced by the interaction of genetic and environmental factors. Epigenetic modifications, especially DNA and RNA methylation, might be the bridge between hereditary and environmental factors. This study aimed to comprehensively analyze the status and prospective trends of the association between T2DM and DNA/RNA methylation modifications by using bibliometric software. Methods All the publications in the Web of Science database for the research of T2DM with DNA and RNA methylation modifications were obtained from the earliest mention to December 2022. CiteSpace software was used to analyze countries, institutions, journals/cited-references, authors/cited-authors, and keywords. Results of the comprehensive visualization and bibliometric analysis were displayed relative to the research hotspots and knowledge structure. Results A total of 1,233 publications related to DNA and RNA methylation modifications and T2DM were collected. The number of publications per year and the overall trend consistently and significantly increased during the investigation period. Based on the highest publication counts, the most influential country was the USA, while Lund University was the most productive institution. DIABETES was considered the most popular journal. The most frequent keywords identified in the field of methylation and T2DM were mainly involved in developmental origin, insulin resistance, and metabolism. The study suggested that the study of methylation modifications had an increasingly significant role in understanding the progression of T2DM. Conclusion CiteSpace visualization software was utilized to investigate the status and trends of DNA and RNA methylation modifications in the pathology of T2DM over the past 30 years. Findings from the study provide a guiding perspective for researchers regarding future research directions in this field.
Collapse
Affiliation(s)
- Cai Jiang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yue Hu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Sinuo Wang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Cong Chen
- Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- *Correspondence: Cong Chen,
| |
Collapse
|
8
|
Zhu D, Li X, Ma J, Zeng J, Gan S, Dong X, Yang J, Lin X, Cai H, Song W, Li X, Zhang K, Zhang Q, Lu Y, Bu R, Shao H, Wang G, Yuan G, Ran X, Liao L, Zhao W, Li P, Sun L, Shi L, Jiang Z, Xue Y, Jiang H, Li Q, Li Z, Fu M, Liang Z, Guo L, Liu M, Xu C, Li W, Yu X, Qin G, Yang Z, Su B, Zeng L, Geng H, Shi Y, Zhao Y, Zhang Y, Yang W, Chen L. Dorzagliatin in drug-naïve patients with type 2 diabetes: a randomized, double-blind, placebo-controlled phase 3 trial. Nat Med 2022; 28:965-973. [PMID: 35551294 DOI: 10.1038/s41591-022-01802-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 03/28/2022] [Indexed: 02/08/2023]
Abstract
Improving glucose sensitivity remains an unmet medical need in treating type 2 diabetes (T2D). Dorzagliatin is a dual-acting, orally bioavailable glucokinase activator that enhances glucokinase activity in a glucose-dependent manner, improves glucose-stimulated insulin secretion and demonstrates effects on glycemic control in patients with T2D. We report the findings of a randomized, double-blind, placebo-controlled phase 3 clinical trial to evaluate the efficacy and safety of dorzagliatin in patients with T2D. Eligible drug-naïve patients with T2D (n = 463) were randomly assigned to the dorzagliatin or placebo group at a ratio of 2:1 for 24 weeks of double-blind treatment, followed by 28 weeks of open-label treatment with dorzagliatin for all patients. The primary efficacy endpoint was the change in glycated hemoglobin from baseline to week 24. Safety was assessed throughout the trial. At week 24, the least-squares mean change in glycated hemoglobin from baseline (95% confidence interval) was -1.07% (-1.19%, -0.95%) in the dorzagliatin group and -0.50% (-0.68%, -0.32%) in the placebo group (estimated treatment difference, -0.57%; 95% confidence interval: -0.79%, -0.36%; P < 0.001). The incidence of adverse events was similar between the two groups. There were no severe hypoglycemia events or drug-related serious adverse events in the dorzagliatin group. In summary, dorzagliatin improved glycemic control in drug-naïve patients with T2D and showed a good tolerability and safety profile.
Collapse
Affiliation(s)
- Dalong Zhu
- Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Xiaoying Li
- Zhongshan Hospital, Fudan University, Shanghai, China
| | | | - Jiao'e Zeng
- Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, China
| | - Shenglian Gan
- The First People's Hospital of Changde City, Changde, China
| | - Xiaolin Dong
- Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jing Yang
- The First Hospital of Shanxi Medical University, Taiyuan, China
| | | | - Hanqing Cai
- The Second Hospital of Jilin University, Changchun, China
| | - Weihong Song
- Chenzhou First People's Hospital, Chenzhou, China
| | - Xuefeng Li
- Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Keqin Zhang
- Tongji Hospital of Tongji University, Shanghai, China
| | - Qiu Zhang
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yibing Lu
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | | | - Huige Shao
- Changsha Central Hospital, Changsha, China
| | - Guixia Wang
- The First Hospital of Jilin University, Changchun, China
| | - Guoyue Yuan
- Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xingwu Ran
- West China Hospital, Sichuan University, Chengdu, China
| | - Lin Liao
- The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Wenjuan Zhao
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ping Li
- Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Li Sun
- Siping Hospital of China Medical University, Siping, China
| | - Lixin Shi
- The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhaoshun Jiang
- The 960th Hospital of the PLA Joint Logistics Support Force, Jinan, China
| | - Yaoming Xue
- Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Hongwei Jiang
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Quanmin Li
- PLA Rocket Force Characteristic Medical Center, Beijing, China
| | | | - Maoxiong Fu
- The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | | | - Lian Guo
- Chongqing University Three Gorges Central Hospital, Chongqing, China
| | - Ming Liu
- Tianjin Medical University General Hospital, Tianjin, China
| | - Chun Xu
- The Third Medical Center of PLA General Hospital, Beijing, China
| | - Wenhui Li
- Peking Union Medical College Hospital, Beijing, China
| | - Xuefeng Yu
- Tongji Hospital, Tongji Medical College of HUST, Wuhan, China
| | - Guijun Qin
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhou Yang
- Jiangxi Pingxiang People's Hospital, Pingxiang, China
| | - Benli Su
- The Second Hospital of Dalian Medical University, Dalian, China
| | - Longyi Zeng
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | | | | - Yu Zhao
- Hua Medicine, Shanghai, China
| | | | - Wenying Yang
- China-Japan Friendship Hospital, Beijing, China.
| | - Li Chen
- Hua Medicine, Shanghai, China.
| |
Collapse
|
9
|
Wang J, Khan SU, Cao P, Chen X, Wang F, Zou D, Li H, Zhao H, Xu K, Jiao D, Yang C, Zhu F, Zhang Y, Su Y, Cheng W, Jia B, Qing Y, Jamal MA, Zhao HY, Wei HJ. Construction of PIK3C3 Transgenic Pig and Its Pathogenesis of Liver Damage. Life (Basel) 2022; 12:630. [PMID: 35629298 PMCID: PMC9146193 DOI: 10.3390/life12050630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/26/2022] [Accepted: 04/08/2022] [Indexed: 11/20/2022] Open
Abstract
As a member of the PIKs family, PIK3C3 participates in autophagy and plays a central role in liver function. Several studies demonstrated that the complete suppression of PIK3C3 in mammals can cause hepatomegaly and hepatosteatosis. However, the function of PIK3C3 overexpression on the liver and other organs is still unknown. In this study, we successfully generated PIK3C3 transgenic pigs through somatic cell nuclear transfer (SCNT) by designing a specific vector for the overexpression of PIK3C3. Plasmid identification was performed through enzyme digestion and transfected into the fetal fibroblasts derived from Diannan miniature pigs. After 2 weeks of culturing, six positive colonies obtained from a total of 14 cell colonies were identified through PCR. One positive cell line was selected as the donor cell line for SCNT for the construction of PIK3C3transgenic pigs. Thirty single blastocysts were collected and identified as PIK3C3 transgenic-positive blastocysts. Two surrogates became pregnant after transferring the reconstructed embryos into four surrogates. Fetal fibroblasts of PIK3C3-positive fetuses identified through PCR were used as donor cells for SCNT to generate PIK3C3 transgenic pigs. To further explore the function of PIK3C3 overexpression, genotyping and phenotyping of the fetuses and piglets obtained were performed by PCR, immunohistochemical, HE, and apoptosis staining. The results showed that inflammatory infiltration and vacuolar formation in hepatocytes and apoptotic cells, and the mRNA expression of NF-κB, TGF-β1, TLR4, TNF-α, and IL-6 significantly increased in the livers of PIK3C3 transgenic pigs when compared with wild-type (WT) pigs. Immunofluorescence staining showed that LC3B and LAMP-1-positive cells increased in the livers of PIK3C3 transgenic pigs. In the EBSS-induced autophagy of the porcine fibroblast cells (PFCs), the accumulated LC3II protein was cleared faster in PIK3C3 transgenic (PFCs) thanWT (PFCs). In conclusion, PIK3C3 overexpression promoted autophagy in the liver and associated molecular mechanisms related to the activation of ULK1, AMBR1, DRAM1, and MTOR, causing liver damage in pigs. Therefore, the construction of PIK3C3 transgenic pigs may provide a new experimental animal resource for liver diseases.
Collapse
Affiliation(s)
- Jing Wang
- Key Laboratory for Porcine Gene Editing and Xenotransplantation in Yunnan Province, Kunming 650201, China; (J.W.); (S.U.K.); (P.C.); (X.C.); (F.W.); (D.Z.); (H.L.); (H.Z.); (K.X.); (D.J.); (C.Y.); (F.Z.); (Y.Z.); (Y.S.); (W.C.); (B.J.); (Y.Q.); (M.A.J.)
- Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Sami Ullah Khan
- Key Laboratory for Porcine Gene Editing and Xenotransplantation in Yunnan Province, Kunming 650201, China; (J.W.); (S.U.K.); (P.C.); (X.C.); (F.W.); (D.Z.); (H.L.); (H.Z.); (K.X.); (D.J.); (C.Y.); (F.Z.); (Y.Z.); (Y.S.); (W.C.); (B.J.); (Y.Q.); (M.A.J.)
- Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Pan Cao
- Key Laboratory for Porcine Gene Editing and Xenotransplantation in Yunnan Province, Kunming 650201, China; (J.W.); (S.U.K.); (P.C.); (X.C.); (F.W.); (D.Z.); (H.L.); (H.Z.); (K.X.); (D.J.); (C.Y.); (F.Z.); (Y.Z.); (Y.S.); (W.C.); (B.J.); (Y.Q.); (M.A.J.)
- Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming 650201, China
| | - Xi Chen
- Key Laboratory for Porcine Gene Editing and Xenotransplantation in Yunnan Province, Kunming 650201, China; (J.W.); (S.U.K.); (P.C.); (X.C.); (F.W.); (D.Z.); (H.L.); (H.Z.); (K.X.); (D.J.); (C.Y.); (F.Z.); (Y.Z.); (Y.S.); (W.C.); (B.J.); (Y.Q.); (M.A.J.)
- Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Fengchong Wang
- Key Laboratory for Porcine Gene Editing and Xenotransplantation in Yunnan Province, Kunming 650201, China; (J.W.); (S.U.K.); (P.C.); (X.C.); (F.W.); (D.Z.); (H.L.); (H.Z.); (K.X.); (D.J.); (C.Y.); (F.Z.); (Y.Z.); (Y.S.); (W.C.); (B.J.); (Y.Q.); (M.A.J.)
- Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Di Zou
- Key Laboratory for Porcine Gene Editing and Xenotransplantation in Yunnan Province, Kunming 650201, China; (J.W.); (S.U.K.); (P.C.); (X.C.); (F.W.); (D.Z.); (H.L.); (H.Z.); (K.X.); (D.J.); (C.Y.); (F.Z.); (Y.Z.); (Y.S.); (W.C.); (B.J.); (Y.Q.); (M.A.J.)
- Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming 650201, China
| | - Honghui Li
- Key Laboratory for Porcine Gene Editing and Xenotransplantation in Yunnan Province, Kunming 650201, China; (J.W.); (S.U.K.); (P.C.); (X.C.); (F.W.); (D.Z.); (H.L.); (H.Z.); (K.X.); (D.J.); (C.Y.); (F.Z.); (Y.Z.); (Y.S.); (W.C.); (B.J.); (Y.Q.); (M.A.J.)
- Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Heng Zhao
- Key Laboratory for Porcine Gene Editing and Xenotransplantation in Yunnan Province, Kunming 650201, China; (J.W.); (S.U.K.); (P.C.); (X.C.); (F.W.); (D.Z.); (H.L.); (H.Z.); (K.X.); (D.J.); (C.Y.); (F.Z.); (Y.Z.); (Y.S.); (W.C.); (B.J.); (Y.Q.); (M.A.J.)
- Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Kaixiang Xu
- Key Laboratory for Porcine Gene Editing and Xenotransplantation in Yunnan Province, Kunming 650201, China; (J.W.); (S.U.K.); (P.C.); (X.C.); (F.W.); (D.Z.); (H.L.); (H.Z.); (K.X.); (D.J.); (C.Y.); (F.Z.); (Y.Z.); (Y.S.); (W.C.); (B.J.); (Y.Q.); (M.A.J.)
- Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Deling Jiao
- Key Laboratory for Porcine Gene Editing and Xenotransplantation in Yunnan Province, Kunming 650201, China; (J.W.); (S.U.K.); (P.C.); (X.C.); (F.W.); (D.Z.); (H.L.); (H.Z.); (K.X.); (D.J.); (C.Y.); (F.Z.); (Y.Z.); (Y.S.); (W.C.); (B.J.); (Y.Q.); (M.A.J.)
- Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Chang Yang
- Key Laboratory for Porcine Gene Editing and Xenotransplantation in Yunnan Province, Kunming 650201, China; (J.W.); (S.U.K.); (P.C.); (X.C.); (F.W.); (D.Z.); (H.L.); (H.Z.); (K.X.); (D.J.); (C.Y.); (F.Z.); (Y.Z.); (Y.S.); (W.C.); (B.J.); (Y.Q.); (M.A.J.)
- Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Feiyan Zhu
- Key Laboratory for Porcine Gene Editing and Xenotransplantation in Yunnan Province, Kunming 650201, China; (J.W.); (S.U.K.); (P.C.); (X.C.); (F.W.); (D.Z.); (H.L.); (H.Z.); (K.X.); (D.J.); (C.Y.); (F.Z.); (Y.Z.); (Y.S.); (W.C.); (B.J.); (Y.Q.); (M.A.J.)
- Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Yaxuan Zhang
- Key Laboratory for Porcine Gene Editing and Xenotransplantation in Yunnan Province, Kunming 650201, China; (J.W.); (S.U.K.); (P.C.); (X.C.); (F.W.); (D.Z.); (H.L.); (H.Z.); (K.X.); (D.J.); (C.Y.); (F.Z.); (Y.Z.); (Y.S.); (W.C.); (B.J.); (Y.Q.); (M.A.J.)
- Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Yanhua Su
- Key Laboratory for Porcine Gene Editing and Xenotransplantation in Yunnan Province, Kunming 650201, China; (J.W.); (S.U.K.); (P.C.); (X.C.); (F.W.); (D.Z.); (H.L.); (H.Z.); (K.X.); (D.J.); (C.Y.); (F.Z.); (Y.Z.); (Y.S.); (W.C.); (B.J.); (Y.Q.); (M.A.J.)
- Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Wenmin Cheng
- Key Laboratory for Porcine Gene Editing and Xenotransplantation in Yunnan Province, Kunming 650201, China; (J.W.); (S.U.K.); (P.C.); (X.C.); (F.W.); (D.Z.); (H.L.); (H.Z.); (K.X.); (D.J.); (C.Y.); (F.Z.); (Y.Z.); (Y.S.); (W.C.); (B.J.); (Y.Q.); (M.A.J.)
- Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Baoyu Jia
- Key Laboratory for Porcine Gene Editing and Xenotransplantation in Yunnan Province, Kunming 650201, China; (J.W.); (S.U.K.); (P.C.); (X.C.); (F.W.); (D.Z.); (H.L.); (H.Z.); (K.X.); (D.J.); (C.Y.); (F.Z.); (Y.Z.); (Y.S.); (W.C.); (B.J.); (Y.Q.); (M.A.J.)
- Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Yubo Qing
- Key Laboratory for Porcine Gene Editing and Xenotransplantation in Yunnan Province, Kunming 650201, China; (J.W.); (S.U.K.); (P.C.); (X.C.); (F.W.); (D.Z.); (H.L.); (H.Z.); (K.X.); (D.J.); (C.Y.); (F.Z.); (Y.Z.); (Y.S.); (W.C.); (B.J.); (Y.Q.); (M.A.J.)
- Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Muhammad Ameen Jamal
- Key Laboratory for Porcine Gene Editing and Xenotransplantation in Yunnan Province, Kunming 650201, China; (J.W.); (S.U.K.); (P.C.); (X.C.); (F.W.); (D.Z.); (H.L.); (H.Z.); (K.X.); (D.J.); (C.Y.); (F.Z.); (Y.Z.); (Y.S.); (W.C.); (B.J.); (Y.Q.); (M.A.J.)
- Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Hong-Ye Zhao
- Key Laboratory for Porcine Gene Editing and Xenotransplantation in Yunnan Province, Kunming 650201, China; (J.W.); (S.U.K.); (P.C.); (X.C.); (F.W.); (D.Z.); (H.L.); (H.Z.); (K.X.); (D.J.); (C.Y.); (F.Z.); (Y.Z.); (Y.S.); (W.C.); (B.J.); (Y.Q.); (M.A.J.)
- Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming 650201, China
| | - Hong-Jiang Wei
- Key Laboratory for Porcine Gene Editing and Xenotransplantation in Yunnan Province, Kunming 650201, China; (J.W.); (S.U.K.); (P.C.); (X.C.); (F.W.); (D.Z.); (H.L.); (H.Z.); (K.X.); (D.J.); (C.Y.); (F.Z.); (Y.Z.); (Y.S.); (W.C.); (B.J.); (Y.Q.); (M.A.J.)
- Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming 650201, China
| |
Collapse
|
10
|
Deng J, Liao Y, Liu J, Liu W, Yan D. Research Progress on Epigenetics of Diabetic Cardiomyopathy in Type 2 Diabetes. Front Cell Dev Biol 2022; 9:777258. [PMID: 35004678 PMCID: PMC8740193 DOI: 10.3389/fcell.2021.777258] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/06/2021] [Indexed: 12/24/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is characterized by diastolic relaxation abnormalities in its initial stages and by clinical heart failure (HF) without dyslipidemia, hypertension, and coronary artery disease in its last stages. DCM contributes to the high mortality and morbidity rates observed in diabetic populations. Diabetes is a polygenic, heritable, and complex condition that is exacerbated by environmental factors. Recent studies have demonstrated that epigenetics directly or indirectly contribute to pathogenesis. While epigenetic mechanisms such as DNA methylation, histone modifications, and non-coding RNAs, have been recognized as key players in the pathogenesis of DCM, some of their impacts remain not well understood. Furthering our understanding of the roles played by epigenetics in DCM will provide novel avenues for DCM therapeutics and prevention strategies.
Collapse
Affiliation(s)
- Jianxin Deng
- Department of Endocrinology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University; Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen, China
| | - Yunxiu Liao
- Health Science Center of Shenzhen University, Shenzhen, China
| | - Jianpin Liu
- Health Science Center of Shenzhen University, Shenzhen, China
| | - Wenjuan Liu
- Health Science Center of Shenzhen University, Shenzhen, China
| | - Dewen Yan
- Department of Endocrinology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University; Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen, China
| |
Collapse
|
11
|
The Relationship between Body Mass Index, Obesity, and LINE-1 Methylation: A Cross-Sectional Study on Women from Southern Italy. DISEASE MARKERS 2021; 2021:9910878. [PMID: 34900031 PMCID: PMC8664509 DOI: 10.1155/2021/9910878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/18/2021] [Accepted: 11/10/2021] [Indexed: 11/17/2022]
Abstract
Uncovering the relationship between body mass index (BMI) and DNA methylation could be useful to understand molecular mechanisms underpinning the effects of obesity. Here, we presented a cross-sectional study, aiming to evaluate the association of BMI and obesity with long interspersed nuclear elements (LINE-1) methylation, among 488 women from Catania, Italy. LINE-1 methylation was assessed in leukocyte DNA by pyrosequencing. We found a negative association between BMI and LINE-1 methylation level in both the unadjusted and adjusted linear regression models. Accordingly, obese women exhibited lower LINE-1 methylation level than their normal weight counterpart. This association was confirmed after adjusting for the effect of age, educational level, employment status, marital status, parity, menopause, and smoking status. Our findings were in line with previous evidence and encouraged further research to investigate the potential role of DNA methylation markers in the management of obesity.
Collapse
|
12
|
Neonatal Vitamin C and Cysteine Deficiencies Program Adult Hepatic Glutathione and Specific Activities of Glucokinase, Phosphofructokinase, and Acetyl-CoA Carboxylase in Guinea Pigs' Livers. Antioxidants (Basel) 2021; 10:antiox10060953. [PMID: 34204849 PMCID: PMC8231532 DOI: 10.3390/antiox10060953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/07/2021] [Accepted: 06/09/2021] [Indexed: 11/17/2022] Open
Abstract
Premature neonates are submitted to an early-life oxidative stress from parenteral nutrition, which is vitamin C (VC) deficient and induces low endogenous levels of glutathione. The oxidative stress caused by these deficiencies may permanently affect liver glycolysis and lipogenesis. This study evaluates the short- and long-term effects of neonatal VC and cysteine deficient diets on redox and energy metabolism. Three-day-old Hartley guinea pigs from both sexes were given a regular or a deficient diet (VC, cysteine, or both) until week 1 of life. Half of the animals were sacrificed at this age, while the other half ate a complete diet until 12 weeks. Liver glutathione and the activity and protein levels of glucokinase, phosphofructokinase, and acetyl-CoA-carboxylase were measured. Statistics: factorial ANOVA (5% threshold). At 1 week, all deficient diets decreased glutathione and the protein levels of glucokinase and phosphofructokinase, while cysteine deficiency decreased acetyl-CoA-carboxylase levels. A similar enzyme level was observed in control animals at 12 weeks. At this age, VC deficiency decreased glutathione, while cysteine increased it. Acetyl-CoA-carboxylase protein levels were increased, which decreased its specific activity. Early-life VC and cysteine deficiencies induce neonatal oxidative stress and an adult-like metabolism, while predisposing to increased lipogenic rates during adulthood.
Collapse
|
13
|
Maugeri A. The Effects of Dietary Interventions on DNA Methylation: Implications for Obesity Management. Int J Mol Sci 2020; 21:ijms21228670. [PMID: 33212948 PMCID: PMC7698434 DOI: 10.3390/ijms21228670] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/28/2020] [Accepted: 11/16/2020] [Indexed: 12/13/2022] Open
Abstract
Previous evidence from in vivo and observational research suggested how dietary factors might affect DNA methylation signatures involved in obesity risk. However, findings from experimental studies are still scarce and, if present, not so clear. The current review summarizes studies investigating the effect of dietary interventions on DNA methylation in the general population and especially in people at risk for or with obesity. Overall, these studies suggest how dietary interventions may induce DNA methylation changes, which in turn are likely related to the risk of obesity and to different response to weight loss programs. These findings might explain the high interindividual variation in weight loss after a dietary intervention, with some people losing a lot of weight while others much less so. However, the interactions between genetic, epigenetic, environmental and lifestyle factors make the whole framework even more complex and further studies are needed to support the hypothesis of personalized interventions against obesity.
Collapse
Affiliation(s)
- Andrea Maugeri
- Department of Medical and Surgical Sciences and Advanced Technologies "GF Ingrassia", University of Catania, 95123 Catania, Italy
| |
Collapse
|
14
|
Guo X, Puttabyatappa M, Domino SE, Padmanabhan V. Developmental programming: Prenatal testosterone-induced changes in epigenetic modulators and gene expression in metabolic tissues of female sheep. Mol Cell Endocrinol 2020; 514:110913. [PMID: 32562712 PMCID: PMC7397566 DOI: 10.1016/j.mce.2020.110913] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/20/2020] [Accepted: 06/08/2020] [Indexed: 12/17/2022]
Abstract
Prenatal testosterone (T)-treated female sheep manifest peripheral insulin resistance and tissue-specific changes in insulin sensitivity with liver and muscle manifesting insulin resistance accompanied by inflammatory, oxidative and lipotoxic state. In contrast, visceral (VAT) and subcutaneous (SAT) adipose tissues are insulin sensitive in spite of VAT manifesting changes in inflammatory and oxidative state. We hypothesized that prenatal T-induced changes in tissue-specific insulin resistance arise from disrupted lipid storage and metabolism gene expression driven by changes in DNA and histone modifying enzymes. Changes in gene expression were assessed in liver, muscle and 4 adipose (VAT, SAT, epicardiac [ECAT] and perirenal [PRAT]) depots collected from control and prenatal T-treated female sheep. Prenatal T-treatment increased lipid droplet and metabolism genes PPARA and PLIN1 in liver, SREBF and PLIN1 in muscle and showed a trend for decrease in PLIN2 in PRAT. Among epigenetic modifying enzymes, prenatal T-treatment increased expression of 1) DNMT1 in liver and DNMT3A in VAT, PRAT, muscle and liver; 2) HDAC1 in ECAT, HDAC2 in muscle with decrease in HDAC3 in VAT; 3) EP300 in VAT and ECAT; and 4) KDM1A in VAT with increases in liver histone acetylation. Increased lipid storage and metabolism genes in liver and muscle are consistent with lipotoxicity in these tissues with increased histone acetylation likely contributing to increased liver PPARA. These findings are suggestive that metabolic defects in prenatal T-treated sheep may arise from changes in key genes mediated, in part, by tissue-specific changes in epigenetic-modifying enzymes.
Collapse
Affiliation(s)
- Xingzi Guo
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA; Department of Obstetrics and Gynecology, 3rd Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China
| | | | - Steven E Domino
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor MI, USA
| | - Vasantha Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor MI, USA.
| |
Collapse
|
15
|
Maugeri A, Barchitta M. How Dietary Factors Affect DNA Methylation: Lesson from Epidemiological Studies. MEDICINA (KAUNAS, LITHUANIA) 2020; 56:E374. [PMID: 32722411 PMCID: PMC7466216 DOI: 10.3390/medicina56080374] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023]
Abstract
Over the past decades, DNA methylation has been proposed as a molecular mechanism underlying the positive or negative effects of diet on human health. Despite the number of studies on this topic is rapidly increasing, the relationship between dietary factors, changes in DNA methylation and health outcomes remains unclear. In this review, we summarize the literature from observational studies (cross-sectional, retrospective, or prospective) which examined the association of dietary factors (nutrients, foods, and dietary patterns) with DNA methylation markers among diseased or healthy people during the lifetime. Next, we discuss the methodological pitfalls by examining strengths and limitations of published studies. Finally, we close with a discussion on future challenges of this field of research, raising the need for large-size prospective studies evaluating the association between diet and DNA methylation in health and diseases for appropriate public health strategies.
Collapse
Affiliation(s)
- Andrea Maugeri
- Department of Medical and Surgical Sciences and Advanced Technologies “GF Ingrassia”, University of Catania, 95123 Catania, Italy;
| | | |
Collapse
|
16
|
Ahmed SAH, Ansari SA, Mensah-Brown EPK, Emerald BS. The role of DNA methylation in the pathogenesis of type 2 diabetes mellitus. Clin Epigenetics 2020; 12:104. [PMID: 32653024 PMCID: PMC7353744 DOI: 10.1186/s13148-020-00896-4] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022] Open
Abstract
Diabetes mellitus (DM) is a chronic condition characterised by β cell dysfunction and persistent hyperglycaemia. The disorder can be due to the absence of adequate pancreatic insulin production or a weak cellular response to insulin signalling. Among the three types of DM, namely, type 1 DM (T1DM), type 2 DM (T2DM), and gestational DM (GDM); T2DM accounts for almost 90% of diabetes cases worldwide. Epigenetic traits are stably heritable phenotypes that result from certain changes that affect gene function without altering the gene sequence. While epigenetic traits are considered reversible modifications, they can be inherited mitotically and meiotically. In addition, epigenetic traits can randomly arise in response to environmental factors or certain genetic mutations or lesions, such as those affecting the enzymes that catalyse the epigenetic modification. In this review, we focus on the role of DNA methylation, a type of epigenetic modification, in the pathogenesis of T2DM.
Collapse
Affiliation(s)
- Sanabil Ali Hassan Ahmed
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al Ain, Abu Dhabi, United Arab Emirates
| | - Suraiya Anjum Ansari
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al Ain, Abu Dhabi, United Arab Emirates
| | - Eric P K Mensah-Brown
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al Ain, Abu Dhabi, United Arab Emirates
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al Ain, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
17
|
Hossan T, Kundu S, Alam SS, Nagarajan S. Epigenetic Modifications Associated with the Pathogenesis of Type 2 Diabetes Mellitus. Endocr Metab Immune Disord Drug Targets 2020; 19:775-786. [PMID: 30827271 DOI: 10.2174/1871530319666190301145545] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/10/2018] [Accepted: 12/28/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND AND OBJECTIVE Type 2 diabetes mellitus (T2DM) is a multifactorial metabolic disorder. Pancreatic β-cell dysfunction and insulin resistance are the most common and crucial events of T2DM. Increasing evidence suggests the association of epigenetic modifications with the pathogenesis of T2DM through the changes in important biological processes including pancreatic β- cell differentiation, development and maintenance of normal β-cell function. Insulin sensitivity by the peripheral glucose uptake tissues is also changed by the altered epigenetic mechanisms. In this review, we discussed the major epigenetic alterations and their effects on β-cell function, insulin secretion and insulin resistance in context of T2DM. METHODS We investigated the presently available epigenetic modifications including DNA methylation, posttranslational histone modifications, ATP-dependent chromatin remodeling and non-coding RNAs related to the pathogenesis of T2DM. Published literatures on this topic were searched both on Google Scholar and Pubmed with related keywords and investigated for relevant information. RESULTS The epigenetic modifications introduce changes in gene expression which are essential for appropriate β-cell development and functions, insulin secretion and sensitivity resulting in the pathogenesis of T2DM. Interestingly, T2DM could also be a prominent reason for the mentioned epigenetic alterations. CONCLUSION This review article emphasized on the epigenetic modifications associated with T2DM and discussed the consequences in deterioration of the disease condition.
Collapse
Affiliation(s)
- Tareq Hossan
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka-1342, Bangladesh
| | - Shoumik Kundu
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka-1342, Bangladesh
| | - Sayeda Sadia Alam
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka-1342, Bangladesh
| | - Sankari Nagarajan
- Cancer Research UK Cambridge Institute (CRUK-CI), University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, United Kingdom
| |
Collapse
|
18
|
Zhang J, Xu W, Han H, Zhang L, Wang T. Dietary Leucine Supplementation Restores Serum Glucose Levels, and Modifying Hepatic Gene Expression Related to the Insulin Signal Pathway in IUGR Piglets. Animals (Basel) 2019; 9:ani9121138. [PMID: 31847151 PMCID: PMC6941017 DOI: 10.3390/ani9121138] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/09/2019] [Accepted: 12/11/2019] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Intrauterine malnutrition may compromise the size and structure of fetal organs and tissues, which leads to lower birth weight and a slower rate of growth after weaning. Intrauterine growth restriction/retardation (IUGR) impairs pancreas function, resulting in the decreased glucose levels in serum. Leucine, one of branched chain amino acids, is an essential amino acid and the substrate of protein synthesis. Leucine also acts as a major regulator of hormone signal transduction, like insulin. Dietary branched chain amino acids or leucine have beneficial effects on the glucose metabolism and glycogen synthesis of muscle. Leucine supplementation improves the insulin sensitivity in liver and muscle and then influences the systemic glucose homeostasis. However, it is still unclear whether leucine supplementation would alter insulin sensitivity in IUGR neonatal piglets. Our results showed that dietary leucine supplementation restored serum glucose concentrations, increased insulin and creatinine concentrations, and enhanced protein kinase adenosine monophosphate-activated γ 3-subunit and glucose transporter type 2 expression. These findings suggest that leucine might play a positive role in hepatic lipid metabolism and glucose metabolism in IUGR. Abstract This study aimed to investigate the effects of leucine with different levels on the insulin resistance in intrauterine growth restriction/retardation (IUGR) piglets. Thirty-two weaned piglets were arranged in a 2 × 2 factorial design and four treatments (n = 8) were as follow: (1) normal weaned piglets fed a basal diet (CONT), (2) IUGR weaned piglets fed a basal diet (IUGR), (3) normal weaned piglets fed a basal diet with the addition of 0.35% l-leucine (C-LEU), and (4) IUGR fed a basal diet with the addition of 0.35% l-leucine (I-LEU) for a 21-days trial. The results showed that compared to the IUGR group, the I-LEU group had higher final body weight and body weight gain, higher serum glucose concentrations, and higher serum insulin concentrations (p < 0.05). The gene expression of phosphatidylinositol 3-kinase p110 gamma, protein kinase adenosine monophosphate-activated γ 3-subunit, glycogen synthase kinase-3 alpha, and glucose transporter type 2 were increased in the I-LEU group as compared to the IUGR group (p < 0.05). It was concluded that dietary leucine supplementation restored serum glucose concentrations, increased insulin and creatinine concentrations, and enhanced protein kinase adenosine monophosphate-activated γ 3-subunit and glucose transporter type 2 expression, suggesting that leucine might play a positive role in hepatic lipid metabolism and glucose metabolism in IUGR.
Collapse
Affiliation(s)
| | | | | | | | - Tian Wang
- Correspondence: ; Tel./Fax: +86-25-84395156
| |
Collapse
|
19
|
Ahmed SM, Johar D, Ali MM, El-Badri N. Insights into the Role of DNA Methylation and Protein Misfolding in Diabetes Mellitus. Endocr Metab Immune Disord Drug Targets 2019; 19:744-753. [DOI: 10.2174/1871530319666190305131813] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/25/2018] [Accepted: 11/29/2018] [Indexed: 12/24/2022]
Abstract
Background:
Diabetes mellitus is a metabolic disorder that is characterized by impaired
glucose tolerance resulting from defects in insulin secretion, insulin action, or both. Epigenetic modifications,
which are defined as inherited changes in gene expression that occur without changes in gene
sequence, are involved in the etiology of diabetes.
Methods:
In this review, we focused on the role of DNA methylation and protein misfolding and their
contribution to the development of both type 1 and type 2 diabetes mellitus.
Results:
Changes in DNA methylation in particular are highly associated with the development of
diabetes. Protein function is dependent on their proper folding in the endoplasmic reticulum. Defective
protein folding and consequently their functions have also been reported to play a role. Early treatment
of diabetes has proven to be of great benefit, as even transient hyperglycemia may lead to pathological
effects and complications later on. This has been explained by the theory of the development of a
metabolic memory in diabetes. The basis for this metabolic memory was attributed to oxidative stress,
chronic inflammation, non-enzymatic glycation of proteins and importantly, epigenetic changes. This
highlights the importance of linking new therapeutics targeting epigenetic mechanisms with traditional
antidiabetic drugs.
Conclusion:
Although new data is evolving on the relation between DNA methylation, protein misfolding,
and the etiology of diabetes, more studies are required for developing new relevant diagnostics
and therapeutics.
Collapse
Affiliation(s)
- Sara M. Ahmed
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, Egypt
| | - Dina Johar
- Biomedical Sciences Program, Zewail City of Science and Technology, Giza, Egypt
| | - Mohamed Medhat Ali
- Biomedical Sciences Program, Zewail City of Science and Technology, Giza, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, Egypt
| |
Collapse
|
20
|
Hunt NJ, Kang SWS, Lockwood GP, Le Couteur DG, Cogger VC. Hallmarks of Aging in the Liver. Comput Struct Biotechnol J 2019; 17:1151-1161. [PMID: 31462971 PMCID: PMC6709368 DOI: 10.1016/j.csbj.2019.07.021] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023] Open
Abstract
While the liver demonstrates remarkable resilience during aging, there is growing evidence that it undergoes all the cellular hallmarks of aging, which increases the risk of liver and systemic disease. The aging process in the liver is driven by alterations of the genome and epigenome that contribute to dysregulation of mitochondrial function and nutrient sensing pathways, leading to cellular senescence and low-grade inflammation. These changes promote multiple phenotypic changes in all liver cells (hepatocytes, liver sinusoidal endothelial, hepatic stellate and Küpffer cells) and impairment of hepatic function. In particular, age-related changes in the liver sinusoidal endothelial cells are a significant but under-recognized risk factor for the development of age-related cardiometabolic disease. Liver aging is driven by transcription and metabolic epigenome alterations. This leads to cellular senescence and low-grade inflammation. Hepatocyte, sinusoidal endothelial, stellate and Küpffer cells undergoes the hallmarks of aging. Each cell type demonstrates phenotypical cellular changes with age.
Collapse
Key Words
- AMPK, 5′ adenosine monophosphate-activated protein kinase
- CR, caloric restriction
- Endothelial
- FOXO, forkhead box O
- Genetic
- HSC, hepatic stellate cell
- Hepatocyte
- IGF-1, insulin like growth factor 1
- IL-6, interleukin 6
- IL-8, interleukin 8
- KC, Küpffer cell
- LSEC, liver sinusoidal endothelial cell
- Mitochondrial dysfunction
- NAD, nicotinamide adenine dinucleotide
- NAFLD, non-alcoholic fatty liver disease
- NO, nitric oxide
- Nutrient sensing pathways
- PDGF, platelet derived growth factor
- PGC-1α, peroxisome proliferator-activated receptor gamma coactivator 1-α
- ROS, reactive oxygen species
- SIRT1, sirtuin 1
- Senescence
- TNFα, tumor necrosis factor alpha
- VEGF, vascular endothelial growth factor
- mTOR, mammalian target of rapamycin
- miR, microRNA
- αSMA, alpha smooth muscle actin
Collapse
Affiliation(s)
- Nicholas J Hunt
- ANZAC Research Institute, Aging and Alzheimer's Institute, Centre for Education and Research on Ageing, Concord Repatriation General Hospital, Concord, NSW, Australia.,The University of Sydney, Concord Clinical School, Sydney Medical School, Sydney, NSW, Australia.,The University of Sydney, Nutrition Ecology, Charles Perkins Centre, Sydney, NSW, Australia
| | - Sun Woo Sophie Kang
- ANZAC Research Institute, Aging and Alzheimer's Institute, Centre for Education and Research on Ageing, Concord Repatriation General Hospital, Concord, NSW, Australia.,The University of Sydney, Nutrition Ecology, Charles Perkins Centre, Sydney, NSW, Australia
| | - Glen P Lockwood
- ANZAC Research Institute, Aging and Alzheimer's Institute, Centre for Education and Research on Ageing, Concord Repatriation General Hospital, Concord, NSW, Australia.,The University of Sydney, Nutrition Ecology, Charles Perkins Centre, Sydney, NSW, Australia
| | - David G Le Couteur
- ANZAC Research Institute, Aging and Alzheimer's Institute, Centre for Education and Research on Ageing, Concord Repatriation General Hospital, Concord, NSW, Australia.,The University of Sydney, Concord Clinical School, Sydney Medical School, Sydney, NSW, Australia.,The University of Sydney, Nutrition Ecology, Charles Perkins Centre, Sydney, NSW, Australia
| | - Victoria C Cogger
- ANZAC Research Institute, Aging and Alzheimer's Institute, Centre for Education and Research on Ageing, Concord Repatriation General Hospital, Concord, NSW, Australia.,The University of Sydney, Concord Clinical School, Sydney Medical School, Sydney, NSW, Australia.,The University of Sydney, Nutrition Ecology, Charles Perkins Centre, Sydney, NSW, Australia
| |
Collapse
|
21
|
Zhou Z, Sun B, Li X, Zhu C. DNA methylation landscapes in the pathogenesis of type 2 diabetes mellitus. Nutr Metab (Lond) 2018; 15:47. [PMID: 29988495 PMCID: PMC6025823 DOI: 10.1186/s12986-018-0283-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 06/18/2018] [Indexed: 01/22/2023] Open
Abstract
Although genetic variations and environmental factors are vital to the development and progression of type 2 diabetes mellitus (T2DM), emerging literature suggest that epigenetics, especially DNA methylation, play a key role in the pathogenesis of T2DM by affecting insulin secretion of pancreatic β cells and the body’s resistance to insulin. Previous studies have elucidated how DNA methylation interacted with various factors in T2DM pathogenesis. This review summarized the role of related methylation genes in insulin-sensitive organs, such as pancreatic islets, skeletal muscle, liver, brain and adipose tissue, as well as peripheral blood cells, comparing the tissue similarity and specificity of methylated genes, aiming at a better understanding of the pathogenesis of T2DM and providing new ideas for the personalized treatment of this metabolism-associated disease.
Collapse
Affiliation(s)
- Zheng Zhou
- 1Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 China
| | - Bao Sun
- 2Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410000 China.,3Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, 410000 China
| | - Xiaoping Li
- 1Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 China
| | - Chunsheng Zhu
- 1Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 China
| |
Collapse
|
22
|
Hu X, Bai T, Xu Z, Liu Q, Zheng Y, Cai L. Pathophysiological Fundamentals of Diabetic Cardiomyopathy. Compr Physiol 2017; 7:693-711. [PMID: 28333387 DOI: 10.1002/cphy.c160021] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Diabetic cardiomyopathy (DCM) was first recognized more than four decades ago and occurred independent of cardiovascular diseases or hypertension in both type 1 and type 2 diabetic patients. The exact mechanisms underlying this disease remain incompletely understood. Several pathophysiological bases responsible for DCM have been proposed, including the presence of hyperglycemia, nonenzymatic glycosylation of large molecules (e.g., proteins), energy metabolic disturbance, mitochondrial damage and dysfunction, impaired calcium handling, reactive oxygen species formation, inflammation, cardiac cell death, and cardiac hypertrophy and fibrosis, leading to impairment of cardiac contractile functions. Increasing evidence also indicates the phenomenon called "metabolic memory" for diabetes-induced cardiovascular complications, for which epigenetic modulation seemed to play an important role, suggesting that the aforementioned pathogenic bases may be regulated by epigenetic modification. Therefore, this review aims at briefly summarizing the current understanding of the pathophysiological bases for DCM. Although how epigenetic mechanisms play a role remains incompletely understood now, extensive clinical and experimental studies have implicated its importance in regulating the cardiac responses to diabetes, which are believed to shed insight into understanding of the pathophysiological and epigenetic mechanisms for the development of DCM and its possible prevention and/or therapy. © 2017 American Physiological Society. Compr Physiol 7:693-711, 2017.
Collapse
Affiliation(s)
- Xinyue Hu
- Center of Cardiovascular Diseases, the First Hospital of Jilin University, Changchun, China.,Pediatric Research Institute at the Department of Pediatrics of the University of Louisville, Louisville, Kentucky, USA
| | - Tao Bai
- Center of Cardiovascular Diseases, the First Hospital of Jilin University, Changchun, China.,Pediatric Research Institute at the Department of Pediatrics of the University of Louisville, Louisville, Kentucky, USA
| | - Zheng Xu
- Center of Cardiovascular Diseases, the First Hospital of Jilin University, Changchun, China.,Pediatric Research Institute at the Department of Pediatrics of the University of Louisville, Louisville, Kentucky, USA
| | - Qiuju Liu
- Department of Hematological Disorders the First Hospital of Jilin University, Changchun, China
| | - Yang Zheng
- Center of Cardiovascular Diseases, the First Hospital of Jilin University, Changchun, China
| | - Lu Cai
- Pediatric Research Institute at the Department of Pediatrics of the University of Louisville, Louisville, Kentucky, USA.,Wendy Novak Diabetes Care Center, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
23
|
Xie C, Wang Q, Wang J, Tan B, Fan Z, Deng ZY, Wu X, Yin Y. Developmental changes in hepatic glucose metabolism in a newborn piglet model: A comparative analysis for suckling period and early weaning period. Biochem Biophys Res Commun 2016; 470:824-30. [PMID: 26802463 DOI: 10.1016/j.bbrc.2016.01.114] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 01/19/2016] [Indexed: 02/07/2023]
Abstract
The liver glucose metabolism, supplying sufficient energy for glucose-dependent tissues, is important in suckling or weaned animals, although there are few studies with piglet model. To better understand the development of glucose metabolism in the piglets during suckling period and early weaning period, we determined the hepatic glycogen content, and investigated the relative protein expression of key enzymes of glucogenesis (GNG) and mRNA levels of some glucose metabolism-related genes. During suckling period, the protein level of G6Pase in the liver of suckling piglets progressively declined with day of age compared with that of newborn piglets (at 1 day of age), whereas the PEPCK level stabilized until day 21 of age, indicating that hepatic GNG capacity gradually weakened in suckling piglets. The synthesis of hepatic glycogen, which was consistent with the fluctuation of glycolytic key genes PFKL and PKLR that gradually decreased after birth and was more or less steady during latter suckling period, although both the mRNA levels of GCK and key glucose transporter GLUT2 presented uptrend in suckling piglets. However, early weaning significantly suppressed the hepatic GNG in the weaned piglets, especially at d 3-5 of weaning period, then gradually recovered at d 7 of weaning period. Meanwhile, PFKL, PKLR and GLUT2 showed the similar trend during weaning period. On the contrast, the hepatic glycogen reached the maximum value when the G6Pase and PEPCK protein expression were at the lowest level, although the GCK level maintained increasing through 7 days of weaning period. Altogether, our study provides evidence that hepatic GNG and glycolysis in newborn piglets were more active than other days during suckling period, and early weaning could significantly suppressed glucose metabolism in liver, but this inhibition would progressively recover at day 7 after weaning.
Collapse
Affiliation(s)
- Chunyan Xie
- Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, China; University of the Chinese Academy of Sciences, Beijing, 10008, China
| | - Qinhua Wang
- Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, China; Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha, 410128, China
| | - Jing Wang
- Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, China; University of the Chinese Academy of Sciences, Beijing, 10008, China
| | - Bie Tan
- Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, China; Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha, 410128, China
| | - Zhiyong Fan
- Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, China; Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha, 410128, China
| | - Ze-yuan Deng
- State Key Laboratory of Food Science and Technology and College of Life Science and Food Engineering, Nanchang University, Nanchang, Jiangxi, 330031, China
| | - Xin Wu
- Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, China; Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha, 410128, China; State Key Laboratory of Food Science and Technology and College of Life Science and Food Engineering, Nanchang University, Nanchang, Jiangxi, 330031, China.
| | - Yulong Yin
- Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, China; Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha, 410128, China; State Key Laboratory of Food Science and Technology and College of Life Science and Food Engineering, Nanchang University, Nanchang, Jiangxi, 330031, China; School of Life Sciences, Hunan Normal University, Changsha, 41008, China; University of the Chinese Academy of Sciences, Beijing, 10008, China.
| |
Collapse
|
24
|
Taki T, Kibayashi K. Characterization of cellular and extracellular DNA in saliva. Leg Med (Tokyo) 2015; 17:471-4. [DOI: 10.1016/j.legalmed.2015.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 10/08/2015] [Accepted: 10/09/2015] [Indexed: 01/08/2023]
|
25
|
Abstract
Colorectal cancer (CRC) is one of most common malignancies and a leading cause of cancer related deaths worldwide. Epigenetic change is an important mechanism of colorectal carcinogenesis. Accumulation of epigenetic changes was found in colorectal cancer and other tumors. Aberrant changes in DNA methylation, histone modification, imprinting, and noncoding RNAs were frequently found in human colorectal cancer. Epigenetic changes may serve as a diagnostic, prognostic, and chemo-sensitive marker. It also becomes a cancer preventive or therapeutic target in some circumstances.
Collapse
Affiliation(s)
- Wenji Yan
- Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, #28 Fuxing Road, Beijing, 100853, China
| | | |
Collapse
|
26
|
|
27
|
Gong YY, Liu YY, Li J, Su L, Yu S, Zhu XN, Cao XP, Xiao HP. Hypermethylation of Cox5a promoter is associated with mitochondrial dysfunction in skeletal muscle of high fat diet-induced insulin resistant rats. PLoS One 2014; 9:e113784. [PMID: 25436770 PMCID: PMC4249960 DOI: 10.1371/journal.pone.0113784] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 10/29/2014] [Indexed: 01/04/2023] Open
Abstract
High-fat diet (HFD) is an environmental factor that contributes to the pathogenesis of obesity and type 2 diabetes. A number of genes influencing oxidative phosphorylation (OXPHOS) were found to be downregulated in skeletal muscle of humans and rats treated with HFD and have been implicated in mitochondrial dysfunction, insulin resistance, and consequent type 2 diabetes. In this study, we hypothesized that DNA methylation plays a crucial role in the regulation of OXPHOS genes in skeletal muscle of rats exposed to HFD. Using whole genome promoter methylation analysis of skeletal muscle followed by qPCR and bisulfite sequencing analysis, we identified hypermethylation of Cox5a in HFD rats. Furthermore, we found that Cox5a hypermethylation was associated with downregulation of Cox5a expression at the mRNA and protein level, and a reduction in mitochondrial complex IV activity and ATP content in HFD-induced insulin resistant rats compared to controls. Moreover, we found that while exposure to palmitate resulted in hypermethylation of the Cox5a promoter in rat myotubes, demethylation with 5-aza-2′-deoxycytidine was associated with preserved Cox5a expression, as well as restoration of complex IV activity and cellular ATP content. These novel observations indicate that Cox5a hypermethylation is associated with mitochondrial dysfunction in skeletal muscle of HFD-induced insulin resistant rats.
Collapse
Affiliation(s)
- Ying-ying Gong
- Department of Geriatrics, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yuan-yuan Liu
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jin Li
- Department of Geriatrics, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Lei Su
- Department of Geriatrics, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shuang Yu
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiao-nan Zhu
- Department of Pharmacology, Zhong-shan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiao-pei Cao
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hai-peng Xiao
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- * E-mail:
| |
Collapse
|
28
|
Epigenetic mechanisms underlying the link between non-alcoholic fatty liver diseases and nutrition. Nutrients 2014; 6:3303-25. [PMID: 25195642 PMCID: PMC4145310 DOI: 10.3390/nu6083303] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 08/06/2014] [Accepted: 08/07/2014] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is defined as a pathologic accumulation of fat in the form of triglycerides (TG) in the liver (steatosis) that is not caused by alcohol. A subgroup of NAFLD patients shows liver cell injury and inflammation coupled with the excessive fat accumulation (steatohepatitis), which is referred to as non-alcoholic steatohepatitis (NASH). Patients with NASH may develop cirrhosis and hepatocellular carcinoma (HCC). NAFLD shares the key features of metabolic syndrome including obesity, hyperlipidemia, hypertension, and insulin resistance. The pathogenesis of NAFLD is multi-factorial, however the oxidative stress seems to plays a major role in the development and progression of the disease. The emerging field of epigenetics provides a new perspective on the pathogenesis of NAFLD. Epigenetics is an inheritable but reversible phenomenon that affects gene expression without altering the DNA sequence and refers to DNA methylation, histone modifications and microRNAs. Epigenetic manipulation through metabolic pathways such as one-carbon metabolism has been proposed as a promising approach to retard the progression of NAFLD. Investigating the epigenetic modifiers in NAFLD may also lead to the development of preventive or therapeutic strategies for NASH-associated complications.
Collapse
|
29
|
F0 maternal BPA exposure induced glucose intolerance of F2 generation through DNA methylation change in Gck. Toxicol Lett 2014; 228:192-9. [DOI: 10.1016/j.toxlet.2014.04.012] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Revised: 04/14/2014] [Accepted: 04/17/2014] [Indexed: 01/03/2023]
|
30
|
Luttmer R, Spijkerman AM, Kok RM, Jakobs C, Blom HJ, Serne EH, Dekker JM, Smulders YM. Metabolic syndrome components are associated with DNA hypomethylation. Obes Res Clin Pract 2014; 7:e106-e115. [PMID: 24331772 DOI: 10.1016/j.orcp.2012.06.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Revised: 05/25/2012] [Accepted: 06/04/2012] [Indexed: 01/22/2023]
Abstract
BACKGROUND Disturbances of DNA methylation have been associated with multiple diseases, including cardiovascular disease, cancer and, as some have suggested, glucometabolic disturbances. Our aim was to assess the association of the metabolic syndrome and its individual components with DNA methylation in a population-based study. MATERIALS AND METHODS In a human population (n = 738) stratified by age, sex and glucose metabolism, we explored associations of the metabolic syndrome according to National Cholesterol Education Program/Adult Treatment Panel-III criteria and its individual components (fasting glucose, high-density lipoprotein cholesterol, triglycerides, blood pressure, waist circumference) with global leukocyte DNA methylation. DNA methylation was measured as the methylcytosine/cytosine ratio in peripheral leukocytes using liquid chromatography-tandem mass spectrometry. RESULTS Individuals with the metabolic syndrome had relative DNA hypomethylation compared to participants without the syndrome (β = -0.05; p = 0.01). This association was mainly attributable to linear associations of two metabolic syndrome components with DNA methylation: fasting plasma glucose (β = -0.02; p = 0.004) and high-density lipoprotein cholesterol (β = 0.07; p = 0.004). People with type 2 diabetes or impaired glucose metabolism had DNA hypomethylation compared to normoglycemic individuals (β = -0.05; p = 0.004). CONCLUSIONS DNA hypomethylation is independently associated with hyperglycemia and low high-density lipoprotein cholesterol, both essential components of the metabolic syndrome. The potential implications and direction of possible causality require further study.
Collapse
Affiliation(s)
- Roosmarijn Luttmer
- Faculty of Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Annemieke M Spijkerman
- Center for Prevention and Health Services Research, National Institute of Public Health and the Environment, Bilthoven, The Netherlands
| | - Robert M Kok
- Department of Clinical Chemistry and Institute for Cardiovascular Research ICaR-VU, VU University Medical Center, Amsterdam, The Netherlands
| | - Carel Jakobs
- Department of Clinical Chemistry and Institute for Cardiovascular Research ICaR-VU, VU University Medical Center, Amsterdam, The Netherlands
| | - Henk J Blom
- Department of Clinical Chemistry and Institute for Cardiovascular Research ICaR-VU, VU University Medical Center, Amsterdam, The Netherlands
| | - Erik H Serne
- Department of Internal Medicine and Institute for Cardiovascular Research ICaR-VU, VU University Medical Center, The Netherlands
| | - Jacqueline M Dekker
- Institute for Research in Extramural Medicine (EMGO Institute), VU University Medical Centre, Amsterdam, The Netherlands
| | - Yvo M Smulders
- Department of Internal Medicine and Institute for Cardiovascular Research ICaR-VU, VU University Medical Center, The Netherlands.
| |
Collapse
|
31
|
Tang L, Ye H, Hong Q, Wang L, Wang Q, Wang H, Xu L, Bu S, Zhang L, Cheng J, Liu P, Le Y, Ye M, Mai Y, Duan S. Elevated CpG island methylation of GCK gene predicts the risk of type 2 diabetes in Chinese males. Gene 2014; 547:329-33. [PMID: 24992032 DOI: 10.1016/j.gene.2014.06.062] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 06/06/2014] [Accepted: 06/27/2014] [Indexed: 12/24/2022]
Abstract
BACKGROUND The GCK gene encodes hexokinase 4, which catalyzes the first step in most glucose metabolism pathways. The purpose of our study is to assess the contribution of GCK methylation to type 2 diabetes (T2D). METHODS AND RESULTS GCK methylation was evaluated in 48 T2D cases and 48 age- and gender-matched controls using the bisulphite pyrosequencing technology. Among the four CpG sites in the methylation assay, CpG4 and the other three CpGs (CpG1-3) were not in high correlation (r<0.5). Significantly elevated methylation levels of GCK CpG4 methylation were observed in T2D patients than in the healthy controls (P=0.004). A breakdown analysis by gender indicated that the association between CpG4 methylation and T2D was specific to males (P=0.002). It is intriguing that another significant male-specific association was also found between GCK CpG4 methylation and total cholesterol (TC) concentration (r=0.304, P=0.036). CONCLUSION Our results showed that elevated GCK CpG4 methylation might suggest a risk of T2D in Chinese males. Gender disparity in GCK CpG4 methylation might provide a clue to elaborate the pathogenesis of T2D.
Collapse
Affiliation(s)
- Linlin Tang
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China; The Affiliated Hospital, School of Medicine, Ningbo University, Ningbo, Zhejiang 315000, China; Diabetes Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Huadan Ye
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Qingxiao Hong
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Lingyan Wang
- Bank of Blood Products, Ningbo No.2 Hospital, Ningbo, Zhejiang 315010, China
| | - Qinwen Wang
- The Affiliated Hospital, School of Medicine, Ningbo University, Ningbo, Zhejiang 315000, China; Diabetes Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Hongwei Wang
- Section of Endocrinology, Pritzker School of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Leiting Xu
- The Affiliated Hospital, School of Medicine, Ningbo University, Ningbo, Zhejiang 315000, China; Diabetes Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Shizhong Bu
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China; Diabetes Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Lina Zhang
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China; Diabetes Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Jia Cheng
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China; Diabetes Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Panpan Liu
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China; Diabetes Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Yanping Le
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China; Diabetes Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Meng Ye
- The Affiliated Hospital, School of Medicine, Ningbo University, Ningbo, Zhejiang 315000, China.
| | - Yifeng Mai
- The Affiliated Hospital, School of Medicine, Ningbo University, Ningbo, Zhejiang 315000, China.
| | - Shiwei Duan
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China; The Affiliated Hospital, School of Medicine, Ningbo University, Ningbo, Zhejiang 315000, China; Diabetes Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China.
| |
Collapse
|
32
|
Abstract
The glucokinase (GK) enzyme (EC 2.7.1.1.) is essential for the use of dietary glucose because it is the first enzyme to phosphorylate glucose in excess in different key tissues such as the pancreas and liver. The objective of the present review is not to fully describe the biochemical characteristics and the genetics of this enzyme but to detail its nutritional regulation in different vertebrates from fish to human. Indeed, the present review will describe the existence of the GK enzyme in different animal species that have naturally different levels of carbohydrate in their diets. Thus, some studies have been performed to analyse the nutritional regulation of the GK enzyme in humans and rodents (having high levels of dietary carbohydrates in their diets), in the chicken (moderate level of carbohydrates in its diet) and rainbow trout (no carbohydrate intake in its diet). All these data illustrate the nutritional importance of the GK enzyme irrespective of feeding habits, even in animals known to poorly use dietary carbohydrates (carnivorous species).
Collapse
|
33
|
Zhu W, Bi M, Liu Y, Wang Y, Pan F, Qiu L, Guo A, Lv H, Yao P, Zhang N, Wang P. Thrombin promotes airway remodeling via protease-activated receptor-1 and transforming growth factor-β1 in ovalbumin-allergic rats. Inhal Toxicol 2014; 25:577-86. [PMID: 23937416 DOI: 10.3109/08958378.2013.813995] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Protease-activated receptor-1 (PAR-1) is widely distributed in platelets and involved in coagulation cascade activated by thrombin. In this study, we intend to explore the role of PAR-1 in the process of thrombin-inducing transforming growth factor-β1 (TGF-β1) to promote airway remodeling in ovalbumin (OVA)-allergic rats. MATERIALS AND METHODS A rat model of chronic asthma was set up by systemic sensitization and repeated challenge to OVA. The doses of thrombin, recombinant hirudin, PAR-1 inhibitor ER-112780-06 varied for different groups. We evaluated the bronchoalveolar lavage fluid (BALF) concentration of thrombin in these groups. The protein and gene expression of PAR-1 was assessed and the expression of TGF-β1 was also detected. RESULTS The PAR-1 mRNA level and the protein level were higher in the airway of asthmatic rats than those of normal rats, and were significantly increased by thrombin treatment but decreased by thrombin-inhibitor treatment. Airway remodeling was strengthened by thrombin but weakened by thrombin inhibitor and PAR-1 antagonist. Expression of TGF-β1 protein in asthmatic rats was significantly increased by thrombin treatment and decreased by thrombin-inhibitor treatment and PAR-1 antagonist treatment. CONCLUSION The expression of PAR-1 is regulated by thrombin that induces the expression of TGF-β1 to promote airway remodeling via PAR-1 in OVA-allergic rats.
Collapse
Affiliation(s)
- Weiwei Zhu
- Department of Pediatrics, Jinan Central Hospital Affiliated to Shandong Univeristy, Jinan, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
GCK gene-body hypomethylation is associated with the risk of coronary heart disease. BIOMED RESEARCH INTERNATIONAL 2014; 2014:151723. [PMID: 24696842 PMCID: PMC3947703 DOI: 10.1155/2014/151723] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 12/31/2013] [Accepted: 01/08/2014] [Indexed: 01/08/2023]
Abstract
Objectives. Glucokinase encoded by GCK is a key enzyme that facilitates phosphorylation of glucose to glucose-6-phosphate. Variants of GCK gene were shown to be associated with type 2 diabetes (T2D) and coronary heart disease (CHD). The goal of this study was to investigate the contribution of GCK gene-body methylation to the risk of CHD. Design and Methods. 36 patients (18 males and 18 females) and 36 age- and sex-matched controls were collected for the current methylation research. DNA methylation level of the CpG island (CGI) region on the GCK gene-body was measured through the sodium bisulfite DNA conversion and pyrosequencing technology. Results. Our results indicated that CHD cases have a much lower methylation level (49.77 ± 6.43%) compared with controls (54.47 ± 7.65%, P = 0.018). In addition, GCK gene-body methylation was found to be positively associated with aging in controls (r = 0.443, P = 0.010). Conclusions. Our study indicated that the hypomethylation of GCK gene-body was significantly associated with the risk of CHD. Aging correlates with an elevation of GCK methylation in healthy controls.
Collapse
|
35
|
Sandoval J, Peiró-Chova L, Pallardó FV, García-Giménez JL. Epigenetic biomarkers in laboratory diagnostics: emerging approaches and opportunities. Expert Rev Mol Diagn 2013; 13:457-71. [PMID: 23782253 DOI: 10.1586/erm.13.37] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Epigenetics has emerged as a new and promising field in recent years. Lifestyle, stress, drugs, physiopathological situations and pharmacological interventions have a great impact on the epigenetic code of the cells by altering the methylome, miRNA expression and the covalent histone modifications. Since there exists a need to find new biomarkers and improve diagnosis for several diseases, the research on epigenetic biomarkers for molecular diagnostics encourages the translation of this field from the bench to clinical practice. In this context, deciphering intricate epigenetic modifications involved in several molecular processes is a challenge that will be solved in the near future. In this review, the authors present an overview of the high-throughput technologies and laboratory techniques available for epigenetic studies, and also discuss which of them are more reliable to be used in a clinical diagnostic laboratory. In addition, the authors describe the most promising epigenetic biomarkers in lung, colorectal and prostate cancer, in which most advances have been achieved. Finally, the authors describe epigenetic biomarkers in some rare diseases; these rare syndromes are paradigms for a specific impaired molecular pathway, thus providing valuable information on the discovery of new epigenetic biomarkers.
Collapse
Affiliation(s)
- Juan Sandoval
- Epigenetics and Cancer Biology, Institut d'Investigació Biomèdica de Bellvitge IDIBELL, Barcelona, Spain
| | | | | | | |
Collapse
|
36
|
Liu HW, Mahmood S, Srinivasan M, Smiraglia DJ, Patel MS. Developmental programming in skeletal muscle in response to overnourishment in the immediate postnatal life in rats. J Nutr Biochem 2013; 24:1859-69. [PMID: 23968580 PMCID: PMC3805821 DOI: 10.1016/j.jnutbio.2013.05.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 05/03/2013] [Indexed: 11/19/2022]
Abstract
Overnourishment during the suckling period [small litter (SL)] results in the development of adult-onset obesity. To investigate the mechanisms that underlie the development of insulin resistance in the skeletal muscle of young and adult female SL rats, the litter size was reduced to 3 female pups/dam (SL) while the control litter had 12 pups/dam from the postnatal Day 3 until Day 21. Protein content, mRNA expression and methylation status of the promoter region of key components in the insulin signaling pathway were determined in the skeletal muscle of SL rats. Overnutrition during the suckling period resulted in increased body weight gains, hyperphagia and adult-onset obesity as well as increased levels of serum insulin, glucose and leptin in SL rats. No differences in the expression of total protein as well as tyrosine phosphorylation of insulin receptor β and glucose transporter 4 (Glut4) were observed in skeletal muscle between two groups at both ages. A significant decrease of total insulin receptor substrate 1 (IRS-1) and an increase in serine phosphorylation of IRS-1 were observed in skeletal muscle from adult SL rats. Hypermethylation of specific cytidyl-3',5'phospho-guanylyl (CpG) dinucleotides in the proximal promoter region was observed for the Irs1 and Glut4 genes, which correlated with the reduction in Irs1 and Glut4 mRNA levels in skeletal muscle of adult SL rats. Our results suggest that epigenetic modifications of the key genes involved in the insulin signaling pathway in skeletal muscle could result in the development of insulin resistance in SL female rats.
Collapse
Affiliation(s)
- Hung-Wen Liu
- Department of Exercise & Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY 14214, USA
| | - Saleh Mahmood
- Department of Biochemistry, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14214, USA
| | - Malathi Srinivasan
- Department of Biochemistry, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14214, USA
| | - Dominic J. Smiraglia
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Mulchand S. Patel
- Department of Biochemistry, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14214, USA
| |
Collapse
|
37
|
Genetic effects on DNA methylation and its potential relevance for obesity in Mexican Americans. PLoS One 2013; 8:e73950. [PMID: 24058506 PMCID: PMC3772804 DOI: 10.1371/journal.pone.0073950] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 07/23/2013] [Indexed: 12/22/2022] Open
Abstract
Several studies have identified effects of genetic variation on DNA methylation patterns and associated heritability, with research primarily focused on Caucasian individuals. In this paper, we examine the evidence for genetic effects on DNA methylation in a Mexican American cohort, a population burdened by a high prevalence of obesity. Using an Illumina-based platform and following stringent quality control procedures, we assessed a total of 395 CpG sites in peripheral blood samples obtained from 183 Mexican American individuals for evidence of heritability, proximal genetic regulation and association with age, sex and obesity measures (i.e. waist circumference and body mass index). We identified 16 CpG sites (∼4%) that were significantly heritable after Bonferroni correction for multiple testing and 27 CpG sites (∼6.9%) that showed evidence of genetic effects. Six CpG sites (∼2%) were associated with age, primarily exhibiting positive relationships, including CpG sites in two genes that have been implicated in previous genome-wide methylation studies of age (FZD9 and MYOD1). In addition, we identified significant associations between three CpG sites (∼1%) and sex, including DNA methylation in CASP6, a gene that may respond to estradiol treatment, and in HSD17B12, which encodes a sex steroid hormone. Although we did not identify any significant associations between DNA methylation and the obesity measures, several nominally significant results were observed in genes related to adipogenesis, obesity, energy homeostasis and glucose homeostasis (ARHGAP9, CDKN2A, FRZB, HOXA5, JAK3, MEST, NPY, PEG3 and SMARCB1). In conclusion, we were able to replicate several findings from previous studies in our Mexican American cohort, supporting an important role for genetic effects on DNA methylation. In addition, we found a significant influence of age and sex on DNA methylation, and report on trend-level, novel associations between DNA methylation and measures of obesity.
Collapse
|
38
|
Ma Y, Xia W, Wang DQ, Wan YJ, Xu B, Chen X, Li YY, Xu SQ. Hepatic DNA methylation modifications in early development of rats resulting from perinatal BPA exposure contribute to insulin resistance in adulthood. Diabetologia 2013; 56:2059-67. [PMID: 23748860 DOI: 10.1007/s00125-013-2944-7] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 05/07/2013] [Indexed: 12/11/2022]
Abstract
AIMS/HYPOTHESIS Perinatal exposure to bisphenol A (BPA), a widely distributed environmental endocrine disruptor, is associated with insulin resistance and diabetes in offspring. The underlying molecular mechanisms could involve epigenetics, as adverse effects induced by environmental exposure in early life are suggested through DNA methylation. In this study we sought to elucidate the relationship between perinatal BPA exposure and alteration of hepatic DNA methylation. METHODS Pregnant Wistar rats were administered BPA (50 μg/kg/day) or corn oil by oral gavage throughout gestation and lactation. Variables associated with insulin resistance and hepatic DNA methylation were examined at postnatal week 3 and week 21 in male offspring. RESULTS In BPA-treated offspring, serum insulin and HOMA-insulin resistance were increased, and the insulin sensitivity index and hepatic glycogen storage were decreased compared with controls at week 21. At week 3, none of these variables were significantly changed. However, hepatic global DNA methylation was decreased, accompanied by overexpression of DNA methyltransferase 3B mRNA at week 3. Meanwhile, perinatal exposure to BPA induced promoter hypermethylation and a reduction in gene expression of hepatic glucokinase. Moreover, increased promoter hypermethylation of Gck became more pronounced in BPA-treated offspring at week 21. CONCLUSIONS/INTERPRETATION Abnormal DNA methylation in hepatic tissue precedes development of insulin resistance induced by perinatal BPA exposure. These findings support the potential role of epigenetics in fetal reprogramming by BPA-induced metabolic disorders.
Collapse
Affiliation(s)
- Y Ma
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Hall E, Dayeh T, Kirkpatrick CL, Wollheim CB, Dekker Nitert M, Ling C. DNA methylation of the glucagon-like peptide 1 receptor (GLP1R) in human pancreatic islets. BMC MEDICAL GENETICS 2013; 14:76. [PMID: 23879380 PMCID: PMC3727960 DOI: 10.1186/1471-2350-14-76] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 07/18/2013] [Indexed: 01/28/2023]
Abstract
BACKGROUND Insulin secretion is enhanced upon the binding of Glucagon-like peptide-1 (GLP-1) to its receptor (GLP1R) in pancreatic β cells. Although a reduced expression of GLP1R in pancreatic islets from type 2 diabetic patients and hyperglycaemic rats has been established, it is still unknown if this is caused by differential DNA methylation of GLP1R in pancreatic islets of type 2 diabetic patients. METHODS In this study, DNA methylation levels of 12 CpG sites close to the transcription start site of GLP1R were analysed in pancreatic islets from 55 non-diabetic and 10 type 2 diabetic human donors as well as in β and α cells isolated from human pancreatic islets. DNA methylation of GLP1R was related to GLP1R expression, HbA1c levels and BMI. Moreover, mRNA expression of MECP2, DNMT1, DNMT3A and DNMT3B was analysed in pancreatic islets of the non-diabetic and type 2 diabetic donors. RESULTS One CpG unit, at position +199 and +205 bp from the transcription start site, showed a small increase in DNA methylation in islets from donors with type 2 diabetes compared to non-diabetic donors (0.53%, p=0.02). Furthermore, DNA methylation levels of one CpG site located 376 bp upstream of the transcription start site of GLP1R correlated negatively with GLP1R expression (rho=-0.34, p=0.008) but positively with BMI and HbA1c (rho=0.30, p=0.02 and rho=0.30, p=0.03, respectively). This specific CpG site is located in an area with known SP1 and SP3 transcription factor binding sites. Moreover, when we compared the DNA methylation of the GLP1R promoter in isolated human β and α cells, we found that it was higher in α- compared with β-cells (p=0.009). Finally, there was a trend towards decreased DNMT3A expression (p=0.056) in type 2 diabetic compared with non-diabetic islets. CONCLUSIONS Together, our study shows that while BMI and HbA1c are positively associated with DNA methylation levels of GLP1R, its expression is negatively associated with DNA methylation of GLP1R in human pancreatic islets.
Collapse
Affiliation(s)
- Elin Hall
- Department of Clinical Sciences, Lund University Diabetes Centre, CRC, Lund University, Scania University Hospital, Malmö, Sweden
| | - Tasnim Dayeh
- Department of Clinical Sciences, Lund University Diabetes Centre, CRC, Lund University, Scania University Hospital, Malmö, Sweden
| | - Clare L Kirkpatrick
- Department of Cell Physiology and Metabolism, University Medical Centre, 1 rue Michel-Servet, 1211, Geneva 4, Switzerland
| | - Claes B Wollheim
- Department of Clinical Sciences, Lund University Diabetes Centre, CRC, Lund University, Scania University Hospital, Malmö, Sweden
- Department of Cell Physiology and Metabolism, University Medical Centre, 1 rue Michel-Servet, 1211, Geneva 4, Switzerland
| | | | - Charlotte Ling
- Department of Clinical Sciences, Lund University Diabetes Centre, CRC, Lund University, Scania University Hospital, Malmö, Sweden
| |
Collapse
|
40
|
Zhang Y, Kent JW, Lee A, Cerjak D, Ali O, Diasio R, Olivier M, Blangero J, Carless MA, Kissebah AH. Fatty acid binding protein 3 (fabp3) is associated with insulin, lipids and cardiovascular phenotypes of the metabolic syndrome through epigenetic modifications in a Northern European family population. BMC Med Genomics 2013; 6:9. [PMID: 23510163 PMCID: PMC3608249 DOI: 10.1186/1755-8794-6-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 03/06/2013] [Indexed: 11/13/2022] Open
Abstract
Background Fatty acid-binding proteins (FABPs) play regulatory roles at the nexus of lipid metabolism and signaling. Dyslipidemia in clinical manifestation frequently co-occurs with obesity, insulin resistance and hypertension in the Metabolic Syndrome (MetS). Animal studies have suggested FABPs play regulatory roles in expressing MetS phenotypes. In our family cohort of Northern European descent, transcript levels in peripheral white blood cells (PWBCs) of a key FABPs, FABP3, is correlated with the MetS leading components. However, evidence supporting the functions of FABPs in humans using genetic approaches has been scarce, suggesting FABPs may be under epigenetic regulation. The objective of this study was to test the hypothesis that CpG methylation status of a key regulator of lipid homeostasis, FABP3, is a quantitative trait associated with status of MetS phenotypes in humans. Methods We used a mass-spec based quantitative method, EpiTYPER®, to profile a CpG island that extends from the promoter to the first exon of the FABP3 gene in our family-based cohort of Northern European descent (n=517). We then conducted statistical analysis of the quantitative relationship of CpG methylation and MetS measures following the variance-component association model. Heritability of each methylation and the effect of age and sex on CpG methylation were also assessed in our families. Results We find that methylation levels of individual CpG units and the regional average are heritable and significantly influenced by age and sex. Regional methylation was strongly associated with plasma total cholesterol (p=0.00028) and suggestively associated with LDL-cholesterol (p=0.00495). Methylation at individual units was significantly associated with insulin sensitivity, lipid particle sizing and diastolic blood pressure (p<0.0028, corrected for multiple testing for each trait). Peripheral white blood cell (PWBC) expression of FABP3 in a separate group of subjects (n=128) negatively correlated with adverse profiles of metabolism (βWHR = −0.72; βLDL-c = −0.53) while positively correlated with plasma adiponectin (β=0.24). Further, we show that differential methylation of FABP3 affects binding activity with nuclear proteins from heart tissue. This region that we found under methylation regulation overlaps with a region actively modified by histone codes in the newly available ENCODE data. Conclusions Our findings suggest that DNA methylation of FABP3 strongly influences MetS, and this may have important implications for cardiovascular disease.
Collapse
Affiliation(s)
- Yi Zhang
- TOPS Obesity and Metabolic Research Center, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Ulrich CM, Toriola AT, Koepl LM, Sandifer T, Poole EM, Duggan C, McTiernan A, Issa JPJ. Metabolic, hormonal and immunological associations with global DNA methylation among postmenopausal women. Epigenetics 2012; 7:1020-8. [PMID: 22869041 DOI: 10.4161/epi.21464] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
DNA methylation is an epigenetic modification essential for the regulation of gene expression that has been implicated in many diseases, including cancer. Few studies have investigated the wide range of potential predictors of global DNA methylation, including biomarkers. Here, we investigated associations between DNA methylation and dietary factors, sex-steroid hormones, metabolic, lipid, inflammation, immune and one-carbon biomarkers. Data and baseline biomarker measurements were obtained from 173 overweight/obese postmenopausal women. Global DNA methylation in lymphocyte DNA was measured using the pyrosequencing assay for LINE-1 repeats. We used correlations and linear regression analyses to investigate associations between continuous data and DNA methylation, while t-tests were used for categorical data. Secondary analyses stratified by serum folate levels and multivitamin use were also conducted. There was little variability in LINE-1 methylation (66.3-79.5%). Mean LINE-1 methylation was significantly higher among women with elevated glucose levels. Mean LINE-1 methylation was also higher among women with high CD4+/CD8+ ratio, and lower among women with elevated vitamin B6, but neither reached statistical significance. In analyses stratified by folate status, DNA methylation was negatively associated with sex hormone concentrations (estrone, estradiol, testosterone and sex hormone binding globulin) among women with low serum folate levels (n = 53). Conversely, among women with high serum folate levels (n = 53), DNA methylation was positively associated with several immune markers (CD4/CD8 ratio, NK1656/lymphocytes and IgA). Results from this screening suggest that global DNA methylation is generally stable, with differential associations for sex hormones and immune markers depending on one-carbon status.
Collapse
Affiliation(s)
- Cornelia M Ulrich
- Division of Preventive Oncology, National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Gilbert ER, Liu D. Epigenetics: the missing link to understanding β-cell dysfunction in the pathogenesis of type 2 diabetes. Epigenetics 2012; 7:841-52. [PMID: 22810088 PMCID: PMC3427279 DOI: 10.4161/epi.21238] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Type 2 diabetes (T2D) is a growing health problem worldwide. While peripheral insulin resistance is common during obesity and aging in both animals and people, progression to T2D is largely due to insulin secretory dysfunction and significant apoptosis of functional β-cells, leading to an inability to compensate for insulin resistance. It is recognized that environmental factors and nutrition play an important role in the pathogenesis of diabetes. However, our knowledge surrounding molecular mechanisms by which these factors trigger β-cell dysfunction and diabetes is still limited. Recent discoveries raise the possibility that epigenetic changes in response to environmental stimuli may play an important role in the development of diabetes. In this paper, we review emerging knowledge regarding epigenetic mechanisms that may be involved in β-cell dysfunction and pathogenesis of diabetes, including the role of nutrition, oxidative stress and inflammation. We will mainly focus on the role of DNA methylation and histone modifications but will also briefly review data on miRNA effects on the pancreatic islets. Further studies aimed at better understanding how epigenetic regulation of gene expression controls β-cell function may reveal potential therapeutic targets for prevention and treatment of diabetes.
Collapse
Affiliation(s)
- Elizabeth R. Gilbert
- Department of Animal and Poultry Sciences; College of Agriculture and Life Sciences; Virginia Tech; Blacksburg, VA USA
| | - Dongmin Liu
- Department of Human Nutrition, Foods and Exercise; College of Agriculture and Life Sciences; Virginia Tech; Blacksburg, VA USA
| |
Collapse
|
43
|
Wang J, Wu Z, Li D, Li N, Dindot SV, Satterfield MC, Bazer FW, Wu G. Nutrition, epigenetics, and metabolic syndrome. Antioxid Redox Signal 2012; 17:282-301. [PMID: 22044276 PMCID: PMC3353821 DOI: 10.1089/ars.2011.4381] [Citation(s) in RCA: 185] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Accepted: 11/01/2011] [Indexed: 01/21/2023]
Abstract
SIGNIFICANCE Epidemiological and animal studies have demonstrated a close link between maternal nutrition and chronic metabolic disease in children and adults. Compelling experimental results also indicate that adverse effects of intrauterine growth restriction on offspring can be carried forward to subsequent generations through covalent modifications of DNA and core histones. RECENT ADVANCES DNA methylation is catalyzed by S-adenosylmethionine-dependent DNA methyltransferases. Methylation, demethylation, acetylation, and deacetylation of histone proteins are performed by histone methyltransferase, histone demethylase, histone acetyltransferase, and histone deacetyltransferase, respectively. Histone activities are also influenced by phosphorylation, ubiquitination, ADP-ribosylation, sumoylation, and glycosylation. Metabolism of amino acids (glycine, histidine, methionine, and serine) and vitamins (B6, B12, and folate) plays a key role in provision of methyl donors for DNA and protein methylation. CRITICAL ISSUES Disruption of epigenetic mechanisms can result in oxidative stress, obesity, insulin resistance, diabetes, and vascular dysfunction in animals and humans. Despite a recognized role for epigenetics in fetal programming of metabolic syndrome, research on therapies is still in its infancy. Possible interventions include: 1) inhibition of DNA methylation, histone deacetylation, and microRNA expression; 2) targeting epigenetically disturbed metabolic pathways; and 3) dietary supplementation with functional amino acids, vitamins, and phytochemicals. FUTURE DIRECTIONS Much work is needed with animal models to understand the basic mechanisms responsible for the roles of specific nutrients in fetal and neonatal programming. Such new knowledge is crucial to design effective therapeutic strategies for preventing and treating metabolic abnormalities in offspring born to mothers with a previous experience of malnutrition.
Collapse
Affiliation(s)
- Junjun Wang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Defa Li
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Ning Li
- State Key Laboratory of AgroBiotechnology, China Agricultural University, Beijing, China
| | - Scott V. Dindot
- Center for Animal Biotechnology and Genomics, Texas A&M University, College Station, Texas
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, Texas
| | - M. Carey Satterfield
- Center for Animal Biotechnology and Genomics, Texas A&M University, College Station, Texas
- Department of Animal Science, Texas A&M University, College Station, Texas
| | - Fuller W. Bazer
- Center for Animal Biotechnology and Genomics, Texas A&M University, College Station, Texas
- Department of Animal Science, Texas A&M University, College Station, Texas
| | - Guoyao Wu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
- Center for Animal Biotechnology and Genomics, Texas A&M University, College Station, Texas
- Department of Animal Science, Texas A&M University, College Station, Texas
| |
Collapse
|
44
|
Chang XX, Yan HM, Xu Q, Xia MF, Bian H, Zhu TF, Gao X. The effects of berberine on hyperhomocysteinemia and hyperlipidemia in rats fed with a long-term high-fat diet. Lipids Health Dis 2012; 11:86. [PMID: 22762542 PMCID: PMC3475055 DOI: 10.1186/1476-511x-11-86] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2012] [Accepted: 06/25/2012] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND The study was undertaken to examine the effects of berberine (BBR) on serum homocysteine, lipids and the aortic lesion in Sprague-Dawley (SD) rats fed with a long-term high-fat diet (HFD). METHODS Healthy male SD rats weighing 190-210 g received randomly standard diet or a high-fat diet for 24 weeks. After 8 weeks of feeding, rats fed with HFD were randomized to receive berberine (200 mg · kg-1· day-1) or vehicle by gavage for 16 weeks. After overnight fasting, all rats were sacrificed and total blood samples were also collected for determinant of fasting serum homocysteine (Hcy), total cholesterol (TC) and low density lipoprotein cholesterol (LDL-c) levels. The aorta was stained with hematoxylin and eosin (HE) and Sudan Ш to evaluate aortic lesion. The livers were dissected out and snap-frozen in liquid nitrogen for hepatic TC content and molecular analysis. 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGR), Lipoprotein receptors and apolipoproteins gene expression in the liver were determined by real-time PCR. RESULTS Intragastrical administration with berberine for 16 weeks lowered serum Hcy in rats fed with a high-fat diet. In parallel, it also decreased body weight and improved serum TC and LDL-c. Berberine also tended to decrease hepatic cholesterol. Consistently, berberine also upregulated LDL receptor (LDLR) mRNA level and suppressed HMGR gene expression. Meanwhile, upon berberine-treated rats, there was a significant increase in apolipoprotein E (apoE) mRNA, but no change in apoAI and scavenger receptor (SR) mRNA in the liver. Further, no atherosclerotic lesions were developed in berberine-treated rats for 16 weeks. CONCLUSION Berberine can counteract HFD-elicited hyperhomocysteinemia and hyperlipidemia partially via upregulating LDLR and apoE mRNA levels and suppressing HMGR gene expression.
Collapse
Affiliation(s)
- Xin-xia Chang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hong-mei Yan
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qiong Xu
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ming-feng Xia
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hua Bian
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Teng-fang Zhu
- Department of Pathology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
45
|
Cianfarani S, Agostoni C, Bedogni G, Berni Canani R, Brambilla P, Nobili V, Pietrobelli A. Effect of intrauterine growth retardation on liver and long-term metabolic risk. Int J Obes (Lond) 2012; 36:1270-7. [PMID: 22531091 DOI: 10.1038/ijo.2012.54] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Intrauterine growth retardation predisposes toward long-term morbidity from type 2 diabetes and cardiovascular disease. To explain this association, the concept of programming was introduced to indicate a process whereby a stimulus or insult at a critical period of development has lasting or lifelong consequences on key endocrine and metabolic pathways. Subtle changes in cell composition of tissues, induced by suboptimal conditions in utero, can influence postnatal physiological functions. There is increasing evidence, suggesting that liver may represent one of the candidate organs targeted by programming, undergoing structural, functional and epigenetic changes following exposure to an unfavorable intrauterine environment. The aim of this review is to provide insights into the molecular mechanisms underlying liver programming that contribute to increase the cardiometabolic risk in subjects with intrauterine growth restriction.
Collapse
Affiliation(s)
- S Cianfarani
- Molecular Endocrinology Unit-DPUO, Bambino Gesù Children's Hospital - 'Rina Balducci' Center of Pediatric Endocrinology, Tor Vergata University, Rome, Italy.
| | | | | | | | | | | | | |
Collapse
|
46
|
|
47
|
Jiang M, Xiang Y, Wang D, Gao J, Liu D, Liu Y, Liu S, Zheng D. Dysregulated expression of miR-146a contributes to age-related dysfunction of macrophages. Aging Cell 2012; 11:29-40. [PMID: 21981419 DOI: 10.1111/j.1474-9726.2011.00757.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Age-associated immune dysfunction, characterized by increased systemic levels of cytokines, manifests as an increased susceptibility to infections. Thus, understanding these negative regulators of the immune response has paved the way to delineating signaling pathways that impact immune senescence. In the present study, we found that miR-146a, which negatively regulated the expression of IL-1β and IL-6, was highly expressed in aged mice. However, there was a lack of response to the stimulation of lipopolysaccharide (LPS) and proinflammatory cytokines in macrophages of aged mice. As a result, the negative feedback regulation loop with miR-146a involving down-regulation of inflammation factors was interrupted in aged mice. Aberrant NF-κB binding to the miR-146a promoter was demonstrated to be associated with the abnormal expression of miR-146a in aged mice. The DNA methyltransferase inhibitor (5-aza-2-deoxycytidine) and the histone deacetylase inhibitor [trichostatin A (TSA)] both significantly up-regulated miR-146a transcriptional activation by altering the DNA-binding activity of NF-κB in macrophages isolated from aged mice, which suggests that DNA methylation and histone acetylation are involved in the suppression of age-dependent miR-146a expression. Additionally, high levels of histone deacetylase (HDACs) expressions contributed to the inhibition of miR-146a expression in LPS-stimulated macrophages from aged mice in vitro. While the suppression of HDACs activities by TSA could improve LPS-induced inflammatory responses owing to up-regulation of miR-146a expression in macrophages from aged mice. These data indicate that the dysregulated expression of miR-146a results in the age-associated dysfunction of macrophages, and miR-146a may be a good target for the treatment of age-related inflammatory diseases.
Collapse
Affiliation(s)
- Minghong Jiang
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Type 2 Diabetes Mellitus (T2DM) is a metabolic disorder influenced by interactions between genetic and environmental factors. Epigenetics conveys specific environmental influences into phenotypic traits through a variety of mechanisms that are often installed in early life, then persist in differentiated tissues with the power to modulate the expression of many genes, although undergoing time-dependent alterations. There is still no evidence that epigenetics contributes significantly to the causes or transmission of T2DM from one generation to another, thus, to the current environment-driven epidemics, but it has become so likely, as pointed out in this paper, that one can expect an efflorescence of epigenetic knowledge about T2DM in times to come.
Collapse
|
49
|
Singh GB, Sharma R, Khullar M. Epigenetics and diabetic cardiomyopathy. Diabetes Res Clin Pract 2011; 94:14-21. [PMID: 21696841 DOI: 10.1016/j.diabres.2011.05.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 05/19/2011] [Accepted: 05/23/2011] [Indexed: 01/21/2023]
Abstract
Cardiovascular complications are a chief cause of mortality and morbidity in diabetic patients. Recent studies suggest that epigenetic changes which may arise as a consequence of environmental factors play an important role in predisposition to disease. Epigenetic mechanisms such as DNA methylation, chromatin remodeling and histone modifications regulate the gene expression in response to environmental signals. Role of epigenetics has been recognized in the pathology of diabetes, however its role in diabetic associated cardiomyopathy remains largely unexplored. In this article, we review current literature on the epigenetic mechanisms involved in diabetes and discuss recent evidence of epigenetic changes that may play an important role in pathophysiology of DCM.
Collapse
Affiliation(s)
- Gurinder Bir Singh
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | | | | |
Collapse
|
50
|
Jiang M, Zhang Y, Liu M, Lan MS, Fei J, Fan W, Gao X, Lu D. Hypermethylation of hepatic glucokinase and L-type pyruvate kinase promoters in high-fat diet-induced obese rats. Endocrinology 2011; 152:1284-9. [PMID: 21239437 DOI: 10.1210/en.2010-1162] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Obesity-dependent insulin resistance and type 2 diabetes mellitus are closely associated with decreased glucose utilization and down-regulation of hepatic glycolytic enzymes expression. Previously, we showed that DNA hypermethylation is involved in age-dependent susceptibility to hepatic insulin resistance and diabetes. However, what we cannot distinguish is whether the age-related obesity contributes to DNA hypermethylation in those natural aging rats. In the present study, we hypothesize that DNA methylation plays a crucial role in the regulation of glycolytic enzymes in the high-fat diet-induced obesity. Here, we report that DNA hypermethylation is correlated with a decline in hepatic glucokinase (Gck) and L-type pyruvate kinase (LPK) expression in high-fat diet-induced obese rats as compared with the control diet group. Down-regulation of Gck and LPK expression are reversed by the 5-aza-2'-deoxycytidine in the cell model of steatosis. These novel observations indicate that DNA methylation is involved in the development of metabolic diseases, such as obesity, insulin resistance, type 2 diabetes mellitus, and nonalcoholic steatohepatitis, suggesting that the hypermethylation level of Gck and LPK promoters may be a useful parameter for the evaluation of obesity-induced insulin resistance and fatty liver.
Collapse
Affiliation(s)
- Minghong Jiang
- State Key Laboratory of Genetic Engineering, Institute of Genetics and Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China 200433
| | | | | | | | | | | | | | | |
Collapse
|