1
|
Yang N, Sun R, Zhang X, Wang J, Wang L, Zhu H, Yuan M, Xu Y, Ge C, He J, Wang M. Alternative pathway of bile acid biosynthesis contributes to ameliorate NASH after induction of NAMPT/NAD +/SIRT1 axis. Biomed Pharmacother 2023; 164:114987. [PMID: 37315437 DOI: 10.1016/j.biopha.2023.114987] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 06/16/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is emerging as a serious liver disorder characterized by hepatic steatosis and liver inflammation. Nicotinamide adenine dinucleotide (NAD+) and NAD+-dependent deacetylase, SIRT1, play important roles in lipid metabolism in non-alcoholic fatty liver disease (NAFLD). However, their effects on liver inflammation and homeostasis of bile acids (BAs), the extensively proved pathophysiological actors in NASH, have not been fully understood. NASH animal model was induced by a methionine-choline-deficient (MCD) diet in C57BL/6J mice and intraperitoneally injected with NAD+ precursor, an agonist of upstream rate-limiting enzyme NAMPT or downstream SIRT1, or their vehicle solvents. Free fatty acid (FFA) was applied to HepG2 cells to construct the cell model. Induction of NAMPT/NAD+/SIRT1 axis could remarkably alleviate the aggravated inflammation in the liver of NASH mice, accompanied by decreased levels of total BAs throughout the enterohepatic system and a switch of BA synthesis from the classic pathway to the alternative pathway, resulting in less production of pro-inflammatory 12-OH BAs. The expressions of key enzymes including cyp7a1, cyp8b1, cyp27a1 and cyp7b1 in BA synthesis were significantly modulated after NAMPT/NAD+/SIRT1 axis induction in both animal and cell models. The levels of pro-inflammatory cytokines in liver were significantly negatively correlated with the intermediates in NAD+ metabolism, which may also be related to their regulation on BA homeostasis. Our results indicated that induction of NAMPT/NAD+/SIRT1 axis may be a potential therapeutic strategy for NASH or its complications related with BAs.
Collapse
Affiliation(s)
- Na Yang
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu, China; Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Runbin Sun
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu, China
| | - Xiaoli Zhang
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Jing Wang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210022, Jiangsu, China
| | - Lulu Wang
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu, China
| | - Huaijun Zhu
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu, China
| | - Man Yuan
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Yifan Xu
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Chun Ge
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China.
| | - Jun He
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China.
| | - Min Wang
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu, China; Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China.
| |
Collapse
|
2
|
Huang Z, Ma Y, Xie Y, Zhao D, Li C. Carrageenan in meat: improvement in lipid metabolism due to Sirtuin1-mediated fatty acid oxidation and inhibited lipid bioavailability. Food Funct 2023. [PMID: 37219362 DOI: 10.1039/d3fo00906h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Kappa-carrageenan (κ-CGN) is widely used in the meat industry. However, its impact on the host metabolism is less revealed. The current study investigated the effect of κ-CGN in pork-based diets on the lipid metabolism of male C57BL/6J mice. The κ-CGN supplement significantly suppressed the increase in body weight by 6.79 g on an average. Supplement of κ-CGN in high-fat diets significantly upregulated the genes and protein expression of Sirtuin1, which was accompanied by the increased gene expression of downstream fatty acids oxidation (Cpt1a and Acadl). The sirtuin1-mediated improvement of lipid metabolism was negatively associated with the levels of bile acids, especially for deoxycholic acid, 3β-cholic acid, glycodeoxycholic acid and glycolithocholic acid. Moreover, κ-CGN in high-fat diets inhibited lipid digestion and absorption, being associated with the decrease in lipid accumulation and improved serum lipid profile. These results highlighted the role of κ-CGN in alleviating diet-induced adiposity by promoting energy expenditure and suppressing the bioavailability of ingested lipids.
Collapse
Affiliation(s)
- Zhiji Huang
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, MOST; Key Laboratory of Meat Processing, MOA; Jiangsu Synergetic Innovation Center of Meat Production, Processing and Quality Control; Nanjing Agricultural University; Nanjing 210095, P.R. China.
| | - Yafang Ma
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, MOST; Key Laboratory of Meat Processing, MOA; Jiangsu Synergetic Innovation Center of Meat Production, Processing and Quality Control; Nanjing Agricultural University; Nanjing 210095, P.R. China.
| | - Yunting Xie
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, MOST; Key Laboratory of Meat Processing, MOA; Jiangsu Synergetic Innovation Center of Meat Production, Processing and Quality Control; Nanjing Agricultural University; Nanjing 210095, P.R. China.
| | - Di Zhao
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, MOST; Key Laboratory of Meat Processing, MOA; Jiangsu Synergetic Innovation Center of Meat Production, Processing and Quality Control; Nanjing Agricultural University; Nanjing 210095, P.R. China.
| | - Chunbao Li
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, MOST; Key Laboratory of Meat Processing, MOA; Jiangsu Synergetic Innovation Center of Meat Production, Processing and Quality Control; Nanjing Agricultural University; Nanjing 210095, P.R. China.
| |
Collapse
|
3
|
Elton AC, Cedarstrom V, Quraishi A, Wuertz B, Murray K, Markowski TW, Seabloom D, Ondrey FG. Metabolic and Metabolomic Effects of Metformin in Murine Model of Pulmonary Adenoma Formation. Nutr Cancer 2023; 75:1014-1027. [PMID: 36688306 DOI: 10.1080/01635581.2023.2165692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Epidemiologic studies of diabetic patients treated with metformin identified significantly lower incidences of cancer. From this, there is growing interest in the use of metformin to treat and prevent cancer. Studies have investigated chemopreventive mechanisms including alterations in calorie intake, cancer metabolism, and cell signaling. Repurposing the drug is challenging due to its metabolic effects and non-uniform effects on different types of cancer. In our previously published studies, we observed that benzo[a]pyrene treated mice receiving metformin significantly reduced lung adenomas; however, mice had reduced weight gain. In this study, we compared chemoprevention diets with and without metformin to evaluate the effects of diet vs. effects of metformin. We also performed tandem mass spectrometry on mouse serum to assess metabolomic alterations associated with metformin treatment. In metformin cohorts, the rate of weight gain was reduced, but weights did not vary between diets. There was no weight difference between diets without metformin. Interestingly, caloric intake was increased in metformin treated mice. Metabolomic analysis revealed metabolite alterations consistent with metformin treatment. Based on these results, we conclude that previous reductions in lung adenomas may have been occurred from anticancer effects of metformin rather than a potentially toxic effect such as calorie restriction.
Collapse
Affiliation(s)
- Andrew C Elton
- Department of Otolaryngology - Head and Neck Surgery, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Vannesa Cedarstrom
- Department of Otolaryngology - Head and Neck Surgery, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Arman Quraishi
- Department of Otolaryngology - Head and Neck Surgery, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Beverly Wuertz
- Department of Otolaryngology - Head and Neck Surgery, University of Minnesota Medical School, Minneapolis, Minnesota, USA.,AeroCore, Department of Otolaryngology - Head and Neck Surgery, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Kevin Murray
- Center for Mass Spectrometry & Proteomics, Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Todd W Markowski
- Center for Mass Spectrometry & Proteomics, Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Donna Seabloom
- Department of Otolaryngology - Head and Neck Surgery, University of Minnesota Medical School, Minneapolis, Minnesota, USA.,AeroCore, Department of Otolaryngology - Head and Neck Surgery, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Frank G Ondrey
- Department of Otolaryngology - Head and Neck Surgery, University of Minnesota Medical School, Minneapolis, Minnesota, USA.,AeroCore, Department of Otolaryngology - Head and Neck Surgery, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|
4
|
Yu C, Li X, Zhao Y, Hu Y. The role of FOXA family transcription factors in glucolipid metabolism and NAFLD. Front Endocrinol (Lausanne) 2023; 14:1081500. [PMID: 36798663 PMCID: PMC9927216 DOI: 10.3389/fendo.2023.1081500] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/17/2023] [Indexed: 02/04/2023] Open
Abstract
Abnormal glucose metabolism and lipid metabolism are common pathological processes in many metabolic diseases, such as nonalcoholic fatty liver disease (NAFLD). Many studies have shown that the forkhead box (FOX) protein subfamily FOXA has a role in regulating glucolipid metabolism and is closely related to hepatic steatosis and NAFLD. FOXA exhibits a wide range of functions ranging from the initiation steps of metabolism such as the development of the corresponding metabolic organs and the differentiation of cells, to multiple pathways of glucolipid metabolism, to end-of-life problems of metabolism such as age-related obesity. The purpose of this article is to review and discuss the currently known targets and signal transduction pathways of FOXA in glucolipid metabolism. To provide more experimental evidence and basis for further research and clinical application of FOXA in the regulation of glucolipid metabolism and the prevention and treatment of NAFLD.
Collapse
Affiliation(s)
- Chuchu Yu
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Institute of Liver Diseases, Shuguang Hospital Affifiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaojing Li
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Institute of Liver Diseases, Shuguang Hospital Affifiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Zhao
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Institute of Liver Diseases, Shuguang Hospital Affifiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Yu Zhao, ; Yiyang Hu,
| | - Yiyang Hu
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Institute of Liver Diseases, Shuguang Hospital Affifiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Clinical Pharmacology, Shuguang Hospital Affifiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Yu Zhao, ; Yiyang Hu,
| |
Collapse
|
5
|
Li T, Yin Y, Mu N, Wang Y, Liu M, Chen M, Jiang W, Yu L, Li Y, Ma H. Metformin-Enhanced Cardiac AMP-Activated Protein Kinase/Atrogin-1 Pathways Inhibit Charged Multivesicular Body Protein 2B Accumulation in Ischemia-Reperfusion Injury. Front Cell Dev Biol 2021; 8:621509. [PMID: 33614629 PMCID: PMC7892907 DOI: 10.3389/fcell.2020.621509] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 12/22/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Cardiac autophagic flux is impaired during myocardial ischemia/reperfusion (MI/R). Impaired autophagic flux may exacerbate MI/R injury. Charged multivesicular body protein 2B (CHMP2B) is a subunit of the endosomal sorting complex required for transport (ESCRT-III) complex that is required for autophagy. However, the reverse role of CHMP2B accumulation in autophagy and MI/R injury has not been established. The objective of this article is to elucidate the roles of AMP-activated protein kinase (AMPK)/atrogin-1 pathways in inhibiting CHMP2B accumulation in ischemia–reperfusion injury. Methods: Male C57BL/6 mice (3–4 months) and H9c2 cardiomyocytes were used to evaluate MI/R and hypoxia/reoxygenation (H/R) injury in vivo and in vitro, respectively. MI/R was built by a left lateral thoracotomy and occluded the left anterior descending artery. H9c2 cells were firstly treated in 95% N2 and 5% CO2 for 15 h and reoxygenation for 1 h. Metformin (100 mg/kg/d) and CHMP2B (Ad-CHMP2B) transfected adenoviruses were administered to the mice. The H9c2 cells were treated with metformin (2.5 mM), MG-132 (10 μM), bafilomycin A1 (10 nM), and compound C (20 μM). Results: Autophagic flux was found to be inhibited in H/R-treated cardiomyocytes and MI/R mice, with elevated cardiac CHMP2B accumulation. Upregulated CHMP2B levels in the in vivo and in vitro experiments were shown to inhibit autophagic flux leading to the deterioration of H/R-cardiomyocytes and MI/R injury. This finding implies that CHMP2B accumulation increases the risk of myocardial ischemia. Metformin suppressed CHMP2B accumulation and ameliorated H/R-induced autophagic dysfunction by activating AMPK. Activated AMPK upregulated the messenger RNA expression and protein levels of atrogin-1, a muscle-specific ubiquitin ligase, in the myocardium. Atrogin-1 significantly enhanced the interaction between atrogin-1 and CHMP2B, therefore, promoting CHMP2B degradation in the MI/R myocardium. Finally, this study revealed that metformin-inhibited CHMP2B accumulation induced autophagic impairment and ischemic susceptibility in vivo through the AMPK-regulated CHMP2B degradation by atrogin-1. Conclusion: Impaired CHMP2B clearance in vitro and in vivo inhibits autophagic flux and weakens the myocardial ischemic tolerance. Metformin treatment degrades CHMP2B through the AMPK-atrogin-1-dependent pathway to maintain the homeostasis of autophagic flux. This is a novel mechanism that enriches the understanding of cardioprotection.
Collapse
Affiliation(s)
- Tian Li
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Yue Yin
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Nan Mu
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Yishi Wang
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Manling Liu
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Mai Chen
- Department of Cardiovascular Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wenhua Jiang
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
| | - Lu Yu
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yan Li
- Department of Cardiovascular Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Heng Ma
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
6
|
Singh P, Reza MI, Syed AA, Garg R, Husain A, Katekar R, Goand UK, Riyazuddin M, Gupta AP, Gayen JR. PSTi8 with metformin ameliorates perimenopause induced steatohepatitis associated ER stress by regulating SIRT-1/SREBP-1c axis. Heliyon 2020; 6:e05826. [PMID: 33426334 PMCID: PMC7779780 DOI: 10.1016/j.heliyon.2020.e05826] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/23/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022] Open
Abstract
Aims Hepatic steatosis in women confronting menopause is the manifestation of substantial fructose consumption and forms a positive feedback loop to develop endoplasmic reticulum (ER) stress. Previously pancreastatin inhibitor peptide-8 (PSTi8) and Metformin (Met) combination effectively ameliorated hepatic lipid accumulation in high fructose diet (HFrD) fed diabetic mice models at reduced doses. Moreover, SIRT-1 plays a crucial role in the regulation of SREBP-1c. Hence we hypothesized that Met and PSTi8 in combination (at therapeutic lower doses) could mitigate hepatic steatosis linked ER stress by activating SIRT-1 and precluding SREBP-1c in HFrD fed 4-Vinylcyclohexenediepoxide (HVCD) induced perimenopausal rats. Main methods HVCD rats were fed HFrD for 12 weeks, accompanied by 14 days of treatment with Met, PSTi8, and combination. We confirmed model establishment by estrus cycle study, estradiol level, and intraperitoneal glucose tolerance test. Plasma lipid profile and liver function were determined. Also, mRNA and protein expressions were examined. Moreover, distribution of SIRT-1 and SREBP-1c was detected in HepG2 cells by immunofluorescence staining. Key findings HVCD group displayed augmented insulin resistance (IR), lipogenesis, and ER stress in the liver. Combination therapy improved the estrus cyclicity, estradiol, and lipid profile of HVCD rats. Met and PSTi8 combination reduced hepatic SREBP-1c and triggered SIRT-1 expression in high fructose-induced insulin-resistant HepG2 cells; consequently, combination therapy attenuated ER stress. Significance Succinctly, present research promotes impetus concerning the remedial impact of Met with PSTi8 at lower therapeutic doses to ameliorate hepatic IR, steatosis, and associated ER stress by revamping the SIRT-1/SREBP-1c axis in perimenopausal rats.
Collapse
Affiliation(s)
- Pragati Singh
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Mohammad Irshad Reza
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Anees A Syed
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Richa Garg
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Athar Husain
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Roshan Katekar
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Umesh K Goand
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mohammed Riyazuddin
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Anand P Gupta
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Jiaur R Gayen
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India.,Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
7
|
García-Calzón S, Perfilyev A, Martinell M, Ustinova M, Kalamajski S, Franks PW, Bacos K, Elbere I, Pihlajamäki J, Volkov P, Vaag A, Groop L, Maziarz M, Klovins J, Ahlqvist E, Ling C. Epigenetic markers associated with metformin response and intolerance in drug-naïve patients with type 2 diabetes. Sci Transl Med 2020; 12:12/561/eaaz1803. [DOI: 10.1126/scitranslmed.aaz1803] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 01/27/2020] [Accepted: 08/24/2020] [Indexed: 12/14/2022]
Abstract
Metformin is the first-line pharmacotherapy for managing type 2 diabetes (T2D). However, many patients with T2D do not respond to or tolerate metformin well. Currently, there are no phenotypes that successfully predict glycemic response to, or tolerance of, metformin. We explored whether blood-based epigenetic markers could discriminate metformin response and tolerance by analyzing genome-wide DNA methylation in drug-naïve patients with T2D at the time of their diagnosis. DNA methylation of 11 and 4 sites differed between glycemic responders/nonresponders and metformin-tolerant/intolerant patients, respectively, in discovery and replication cohorts. Greater methylation at these sites associated with a higher risk of not responding to or not tolerating metformin with odds ratios between 1.43 and 3.09 per 1-SD methylation increase. Methylation risk scores (MRSs) of the 11 identified sites differed between glycemic responders and nonresponders with areas under the curve (AUCs) of 0.80 to 0.98. MRSs of the 4 sites associated with future metformin intolerance generated AUCs of 0.85 to 0.93. Some of these blood-based methylation markers mirrored the epigenetic pattern in adipose tissue, a key tissue in diabetes pathogenesis, and genes to which these markers were annotated to had biological functions in hepatocytes that altered metformin-related phenotypes. Overall, we could discriminate between glycemic responders/nonresponders and participants tolerant/intolerant to metformin at diagnosis by measuring blood-based epigenetic markers in drug-naïve patients with T2D. This epigenetics-based tool may be further developed to help patients with T2D receive optimal therapy.
Collapse
Affiliation(s)
- Sonia García-Calzón
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Scania University Hospital, 214 28 Malmö, Sweden
- Department of Nutrition, Food Science and Physiology, University of Navarra, 31008 Pamplona, Spain
| | - Alexander Perfilyev
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Scania University Hospital, 214 28 Malmö, Sweden
| | - Mats Martinell
- Department of Public Health and Caring Sciences, Uppsala University, 751 22 Uppsala, Sweden
| | - Monta Ustinova
- Latvian Biomedical Research and Study Centre, Rātsupītes Street 1, k-1, Riga LV-1067, Latvia
| | - Sebastian Kalamajski
- Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University Diabetes Centre, 214 28 Malmö, Sweden
| | - Paul W. Franks
- Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University Diabetes Centre, 214 28 Malmö, Sweden
| | - Karl Bacos
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Scania University Hospital, 214 28 Malmö, Sweden
| | - Ilze Elbere
- Latvian Biomedical Research and Study Centre, Rātsupītes Street 1, k-1, Riga LV-1067, Latvia
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, Internal Medicine, University of Eastern Finland, 70211 Kuopio, Finland
- Clinical Nutrition and Obesity Center, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Petr Volkov
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Scania University Hospital, 214 28 Malmö, Sweden
| | - Allan Vaag
- Type 2 Diabetes Biology Research, Steno Diabetes Center, 2820 Gentofte, Denmark
| | - Leif Groop
- Genomics, Diabetes and Endocrinology Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Scania University Hospital, 214 28 Malmö, Sweden
| | - Marlena Maziarz
- Bioinformatics Unit, Department of Clinical Sciences, Lund University Diabetes Centre, 214 28 Malmö, Sweden
| | - Janis Klovins
- Latvian Biomedical Research and Study Centre, Rātsupītes Street 1, k-1, Riga LV-1067, Latvia
- Faculty of Biology, University of Latvia, Riga LV-1004, Latvia
| | - Emma Ahlqvist
- Genomics, Diabetes and Endocrinology Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Scania University Hospital, 214 28 Malmö, Sweden
| | - Charlotte Ling
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Scania University Hospital, 214 28 Malmö, Sweden
| |
Collapse
|
8
|
Qin X, Du D, Chen Q, Wu M, Wu T, Wen J, Jin Y, Zhang J, Wang S. Metformin prevents murine ovarian aging. Aging (Albany NY) 2020; 11:3785-3794. [PMID: 31182682 PMCID: PMC6594816 DOI: 10.18632/aging.102016] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 05/31/2019] [Indexed: 02/07/2023]
Abstract
A number of studies have shown that metformin can delay aging process and extend healthy lifespan in animals. However, its role in female reproductive lifespan is unclear. This study was aimed to explore the potential anti-aging effect of metformin on the ovary and its possible mechanisms. Female C57BL/6 mice of 27-week old were divided into two groups, the control group (CON) and metformin-treated group (MET). CON mice were fed ad libitum, while MET mice were fed on chows supplied with 100mg/kg metformin for half a year. Ovarian reserve and function were assessed by ovarian follicle counts, estrous cycle and sex hormones levels. The expressions of oxidized metabolites, such as 8-hydroxy-2´-deoxyguanosine (8-OHdG), 4-hydroxynonenal (4-HNE), nitrotyrosine (NTY), and ovarian aging associated proteins P16, SIRT1, p-rpS6 and Bcl2 were examined. The MET mice exhibited increased level of serum E2 hormone and higher percentage of regular estrous cycles after 6 months' feeding, compared to the CON mice. The amount of primordial and primary follicles and the expression of SIRT1 were significantly increased, but the levels of P16, 8-OHdG, 4-HNE and p-rpS6 were decreased in the MET mice. These results indicate that metformin can delay ovarian aging process, probably by inducing the expression of SIRT1 and reducing the oxidative damage.
Collapse
Affiliation(s)
- Xian Qin
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.,Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Dingfu Du
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Qian Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Tong Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Jingyi Wen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yan Jin
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Jinjin Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
9
|
Garzel B, Hu T, Li L, Lu Y, Heyward S, Polli J, Zhang L, Huang SM, Raufman JP, Wang H. Metformin Disrupts Bile Acid Efflux by Repressing Bile Salt Export Pump Expression. Pharm Res 2020; 37:26. [PMID: 31907698 DOI: 10.1007/s11095-019-2753-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/26/2019] [Indexed: 12/12/2022]
Abstract
PURPOSE The bile salt export pump (BSEP), a key player in hepatic bile acid clearance, has been the center of research on drug-induced cholestasis. However, such studies focus primarily on the direct inhibition of BSEP, often overlooking the potential impact of transcriptional repression. This work aims to explore the disruption of bile acid efflux caused by drug-induced BSEP repression. METHODS BSEP activity was analyzed in human primary hepatocytes (HPH) using a traditional biliary-clearance experiment and a modified efflux assay, which includes a 72-h pretreatment prior to efflux measurement. Relative mRNA and protein expressions were examined by RT-PCR and Western blotting, respectively. RESULTS Metformin concentration-dependently repressed BSEP expression in HPH. Although metformin did not directly inhibit BSEP activity, longer metformin exposure reduced BSEP transport function in HPH by down-regulating BSEP expression. BSEP repression by metformin was found to be AMP-activated protein kinase-independent. Additional screening of 10 reported cholestatic non-BSEP inhibitors revealed that the anti-cancer drug tamoxifen also markedly repressed BSEP expression and reduced BSEP activity in HPH. CONCLUSIONS Repression of BSEP alone is sufficient to disrupt hepatic bile acid efflux. Metformin and tamoxifen appear to be prototypes of a class of BSEP repressors that may cause drug-induced cholestasis through gene repression instead of direct BSEP inhibition.
Collapse
Affiliation(s)
- Brandy Garzel
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland, 21201, USA
| | - Tao Hu
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland, 21201, USA
| | - Linhao Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland, 21201, USA
| | - Yuanfu Lu
- Key Laboratory of Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Scott Heyward
- BioIVT, 1450 S Rolling Road, Baltimore, Maryland, 21227, USA
| | - James Polli
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland, 21201, USA
| | - Lei Zhang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Shiew-Mei Huang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Jean-Pierre Raufman
- Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, 22 South Greene Street, Baltimore, Maryland, 21201, USA.,VA Maryland Health Care System, 10 N. Greene Street, Baltimore, Maryland, 21201, USA
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland, 21201, USA.
| |
Collapse
|
10
|
Dollerup OL, Trammell SAJ, Hartmann B, Holst JJ, Christensen B, Møller N, Gillum MP, Treebak JT, Jessen N. Effects of Nicotinamide Riboside on Endocrine Pancreatic Function and Incretin Hormones in Nondiabetic Men With Obesity. J Clin Endocrinol Metab 2019; 104:5703-5714. [PMID: 31390002 DOI: 10.1210/jc.2019-01081] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 08/01/2019] [Indexed: 01/14/2023]
Abstract
OBJECTIVE Augmenting nicotinamide adenine dinucleotide (NAD+) metabolism through dietary provision of NAD+ precursor vitamins translates to improved glucose handling in rodent models of obesity and diabetes. Preclinical evidence suggests that the NAD+/SIRT1 axis may be implicated in modulating important gut-related aspects of glucose regulation. We sought to test whether NAD+ precursor supplementation with nicotinamide riboside (NR) affects β-cell function, α-cell function, and incretin hormone secretion as well as circulating bile acid levels in humans. DESIGN A 12-week randomized, double-blind, placebo-controlled, parallel-group trial in 40 males with obesity and insulin resistance allocated to NR at 1000 mg twice daily (n = 20) or placebo (n = 20). Two-hour 75-g oral glucose tolerance tests were performed before and after the intervention, and plasma concentrations of glucose, insulin, C-peptide, glucagon, glucagon-like peptide 1 (GLP-1), and glucose-dependent insulinotropic polypeptide (GIP) were determined. β-Cell function indices were calculated based on glucose, insulin, and C-peptide measurements. Fasting plasma concentrations of bile acids were determined. RESULTS NR supplementation during 12 weeks did not affect fasting or postglucose challenge concentrations of glucose, insulin, C-peptide, glucagon, GLP-1, or GIP, and β-cell function did not respond to the intervention. Additionally, no changes in circulating adipsin or bile acids were observed following NR supplementation. CONCLUSION The current study does not provide evidence to support that dietary supplementation with the NAD+ precursor NR serves to impact glucose tolerance, β-cell secretory capacity, α-cell function, and incretin hormone secretion in nondiabetic males with obesity. Moreover, bile acid levels in plasma did not change in response to NR supplementation.
Collapse
Affiliation(s)
- Ole L Dollerup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Medical Research Laboratory, Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark
| | - Samuel A J Trammell
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bolette Hartmann
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Section for Translational Metabolic Physiology, University of Copenhagen, Copenhagen Denmark
| | - Jens J Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Section for Translational Metabolic Physiology, University of Copenhagen, Copenhagen Denmark
| | - Britt Christensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Medical Research Laboratory, Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Niels Møller
- Medical Research Laboratory, Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Endocrinology, Aarhus University Hospital, Aarhus, Denmark
| | - Matthew P Gillum
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Jessen
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
11
|
Huang J, Shen G, Ren H, Zhang Z, Yu X, Zhao W, Shang Q, Cui J, Yu P, Peng J, Liang D, Yang Z, Jiang X. Role of forkhead box gene family in bone metabolism. J Cell Physiol 2019; 235:1986-1994. [DOI: 10.1002/jcp.29178] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 08/23/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Jinjing Huang
- Guangzhou University of Chinese Medicine Guangzhou China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
| | - Gengyang Shen
- Guangzhou University of Chinese Medicine Guangzhou China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
| | - Hui Ren
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
- Department of Spinal Surgery The First Affiliated Hospital of Guangzhou University of Chinese Medicine Guangzhou China
| | - Zhida Zhang
- Guangzhou University of Chinese Medicine Guangzhou China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
| | - Xiang Yu
- Guangzhou University of Chinese Medicine Guangzhou China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
| | - Wenhua Zhao
- Guangzhou University of Chinese Medicine Guangzhou China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
| | - Qi Shang
- Guangzhou University of Chinese Medicine Guangzhou China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
| | - Jianchao Cui
- Guangzhou University of Chinese Medicine Guangzhou China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
| | - Peiyuan Yu
- Guangzhou University of Chinese Medicine Guangzhou China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
| | - Jiancheng Peng
- Guangzhou University of Chinese Medicine Guangzhou China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
| | - De Liang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
- Department of Spinal Surgery The First Affiliated Hospital of Guangzhou University of Chinese Medicine Guangzhou China
| | - Zhidong Yang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
- Department of Spinal Surgery The First Affiliated Hospital of Guangzhou University of Chinese Medicine Guangzhou China
| | - Xiaobing Jiang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
- Department of Spinal Surgery The First Affiliated Hospital of Guangzhou University of Chinese Medicine Guangzhou China
| |
Collapse
|
12
|
Park JH, Kim YH, Park EH, Lee SJ, Kim H, Kim A, Lee SB, Shim S, Jang H, Myung JK, Park S, Lee SJ, Kim MJ. Effects of metformin and phenformin on apoptosis and epithelial-mesenchymal transition in chemoresistant rectal cancer. Cancer Sci 2019; 110:2834-2845. [PMID: 31278880 PMCID: PMC6726705 DOI: 10.1111/cas.14124] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 12/13/2022] Open
Abstract
Recurrence and chemoresistance in colorectal cancer remain important issues for patients treated with conventional therapeutics. Metformin and phenformin, previously used in the treatment of diabetes, have been shown to have anticancer effects in various cancers, including breast, lung and prostate cancers. However, their molecular mechanisms are still unclear. In this study, we examined the effects of these drugs in chemoresistant rectal cancer cell lines. We found that SW837 and SW1463 rectal cancer cells were more resistant to ionizing radiation and 5‐fluorouracil than HCT116 and LS513 colon cancer cells. In addition, metformin and phenformin increased the sensitivity of these cell lines by inhibiting cell proliferation, suppressing clonogenic ability and increasing apoptotic cell death in rectal cancer cells. Signal transducer and activator of transcription 3 and transforming growth factor‐β/Smad signaling pathways were more activated in rectal cancer cells, and inhibition of signal transducer and activator of transcription 3 expression using an inhibitor or siRNA sensitized rectal cancer cells to chemoresistant by inhibition of the expression of antiapoptotic proteins, such as X‐linked inhibitor of apoptosis, survivin and cellular inhibitor of apoptosis protein 1. Moreover, metformin and phenformin inhibited cell migration and invasion by suppression of transforming growth factor β receptor 2‐mediated Snail and Twist expression in rectal cancer cells. Therefore, metformin and phenformin may represent a novel strategy for the treatment of chemoresistant rectal cancer by targeting signal transducer and activator of transcription 3 and transforming growth factor‐β/Smad signaling.
Collapse
Affiliation(s)
- Ji-Hye Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Young-Heon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Eun Hyeh Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Sun-Joo Lee
- Laboratory of Experimental Pathology, Department of Pathology, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Hyewon Kim
- Laboratory of Experimental Pathology, Department of Pathology, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Areumnuri Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Seung Bum Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Sehwan Shim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Hyosun Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Jae Kyung Myung
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea.,Laboratory of Experimental Pathology, Department of Pathology, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Sunhoo Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea.,Laboratory of Experimental Pathology, Department of Pathology, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Su-Jae Lee
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Min Jung Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| |
Collapse
|
13
|
Ye C, Chen M, Chen E, Li W, Wang S, Ding Q, Wang C, Zhou C, Tang L, Hou W, Hang K, He R, Pan Z, Zhang W. Knockdown of FOXA2 enhances the osteogenic differentiation of bone marrow-derived mesenchymal stem cells partly via activation of the ERK signalling pathway. Cell Death Dis 2018; 9:836. [PMID: 30082727 PMCID: PMC6079048 DOI: 10.1038/s41419-018-0857-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/26/2018] [Accepted: 07/05/2018] [Indexed: 02/07/2023]
Abstract
Forkhead box protein A2 (FOXA2) is a core transcription factor that controls cell differentiation and may have an important role in bone metabolism. However, the role of FOXA2 during osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) remains largely unknown. In this study, decreased expression of FOXA2 was observed during osteogenic differentiation of rat BMSCs (rBMSCs). FOXA2 knockdown significantly increased osteoblast-specific gene expression, the number of mineral deposits and alkaline phosphatase activity, whereas FOXA2 overexpression inhibited osteogenesis-specific activities. Moreover, extracellular signal-regulated protein kinase (ERK) signalling was upregulated following knockdown of FOXA2. The enhanced osteogenesis due to FOXA2 knockdown was partially rescued by an ERK inhibitor. Using a rat tibial defect model, a rBMSC sheet containing knocked down FOXA2 significantly improved bone healing. Collectively, these findings indicated that FOXA2 had an essential role in osteogenic differentiation of BMSCs, partly by activation of the ERK signalling pathway.
Collapse
Affiliation(s)
- Chenyi Ye
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China
| | - Mo Chen
- Department of Rheumatology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Erman Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China
| | - Weixu Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China
| | - Shengdong Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China
| | - Qianhai Ding
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China
| | - Cong Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China
| | - Chenhe Zhou
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China
| | - Lan Tang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China
| | - Weiduo Hou
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China
| | - Kai Hang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China.,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China
| | - Rongxin He
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China. .,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China.
| | - Zhijun Pan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China. .,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China.
| | - Wei Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China. .,Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China.
| |
Collapse
|
14
|
Sillner N, Walker A, Koch W, Witting M, Schmitt-Kopplin P. Metformin impacts cecal bile acid profiles in mice. J Chromatogr B Analyt Technol Biomed Life Sci 2018. [PMID: 29522956 DOI: 10.1016/j.jchromb.2018.02.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bile acids (BAs) are major components of bile synthesized from cholesterol and take part in the digestion of dietary lipids, as well as having signaling functions. They undergo extensive microbial metabolism inside the gastrointestinal tract. Here, we present a method of ultra-high pressure liquid chromatography coupled to ion trap mass spectrometry for quantification of 45 BAs in mouse cecum. The system was validated in regard to sensitivity with limits of detection and quantification (0.6-24.9 nM), interday accuracy (102.4%), interday precision (15.2%), recovery rate (74.7%), matrix effect (98.2%) and carry-over effect (<1.1%). Afterwards, we applied our method to investigate the effect of metformin on BA profiles. Diabetic mice were treated with metformin for 1 day or 14 days. One day of treatment resulted in a significant increase of total BA concentration (2.7-fold increase; db/db metformin 5.32 μmol/g, db/db control mice 1.95 μmol/g), most notable in levels of 7-oxodeoxycholic, 3-dehydrocholic and cholic acid. We observed only minor impact on BA metabolism after 14 days of metformin treatment, compared to the single treatment. Furthermore, healthy wild type mice had elevated concentrations of allocholic and ω-muricholic acid compared to diabetic mice. Our method proved the applicability of profiling BAs in cecum to investigate intestinal BA metabolism in diabetes and pharmacological applications.
Collapse
Affiliation(s)
- Nina Sillner
- ZIEL Institute for Food and Health, Technical University of Munich, Freising, Germany; Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, Neuherberg, Germany
| | - Alesia Walker
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, Neuherberg, Germany.
| | - Wendelin Koch
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, Neuherberg, Germany
| | - Michael Witting
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, Neuherberg, Germany; Chair of Analytical Food Chemistry, Technical University of Munich, Freising, Germany
| | - Philippe Schmitt-Kopplin
- ZIEL Institute for Food and Health, Technical University of Munich, Freising, Germany; Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, Neuherberg, Germany; Chair of Analytical Food Chemistry, Technical University of Munich, Freising, Germany
| |
Collapse
|
15
|
Aliper A, Jellen L, Cortese F, Artemov A, Karpinsky-Semper D, Moskalev A, Swick AG, Zhavoronkov A. Towards natural mimetics of metformin and rapamycin. Aging (Albany NY) 2017; 9:2245-2268. [PMID: 29165314 PMCID: PMC5723685 DOI: 10.18632/aging.101319] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 11/02/2017] [Indexed: 12/14/2022]
Abstract
Aging is now at the forefront of major challenges faced globally, creating an immediate need for safe, widescale interventions to reduce the burden of chronic disease and extend human healthspan. Metformin and rapamycin are two FDA-approved mTOR inhibitors proposed for this purpose, exhibiting significant anti-cancer and anti-aging properties beyond their current clinical applications. However, each faces issues with approval for off-label, prophylactic use due to adverse effects. Here, we initiate an effort to identify nutraceuticals-safer, naturally-occurring compounds-that mimic the anti-aging effects of metformin and rapamycin without adverse effects. We applied several bioinformatic approaches and deep learning methods to the Library of Integrated Network-based Cellular Signatures (LINCS) dataset to map the gene- and pathway-level signatures of metformin and rapamycin and screen for matches among over 800 natural compounds. We then predicted the safety of each compound with an ensemble of deep neural network classifiers. The analysis revealed many novel candidate metformin and rapamycin mimetics, including allantoin and ginsenoside (metformin), epigallocatechin gallate and isoliquiritigenin (rapamycin), and withaferin A (both). Four relatively unexplored compounds also scored well with rapamycin. This work revealed promising candidates for future experimental validation while demonstrating the applications of powerful screening methods for this and similar endeavors.
Collapse
Affiliation(s)
- Alexander Aliper
- Insilico Medicine, Inc, Research Department, Baltimore, MD 21218, USA
| | - Leslie Jellen
- Insilico Medicine, Inc, Research Department, Baltimore, MD 21218, USA
| | - Franco Cortese
- Biogerontology Research Foundation, Research Department, Oxford, United Kingdom
- Department of Biomedical and Molecular Science, Queen's University School of Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Artem Artemov
- Insilico Medicine, Inc, Research Department, Baltimore, MD 21218, USA
| | | | - Alexey Moskalev
- Laboratory of Molecular Radiobiology and Gerontology, Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences, Syktyvkar, 167982, Russia
| | | | - Alex Zhavoronkov
- Insilico Medicine, Inc, Research Department, Baltimore, MD 21218, USA
- Biogerontology Research Foundation, Research Department, Oxford, United Kingdom
| |
Collapse
|