1
|
Ponte ME, Prom JC, Newcomb MA, Jordan AB, Comfort LL, Hu J, Puchalska P, Geisler CE, Hayes MR, Morris EM. Reduced Liver Mitochondrial Energy Metabolism Impairs Food Intake Regulation Following Gastric Preloads and Fasting. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.24.620086. [PMID: 39554188 PMCID: PMC11565831 DOI: 10.1101/2024.10.24.620086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
The capacity of the liver to serve as a peripheral sensor in the regulation of food intake has been debated for over half a century. The anatomical position and physiological roles of the liver suggest it is a prime candidate to serve as an interoceptive sensor of peripheral tissue and systemic energy state. Importantly, maintenance of liver ATP levels and within-meal food intake inhibition is impaired in human subjects with obesity and obese pre-clinical models. We demonstrate that decreased hepatic mitochondrial energy metabolism in liver-specific, heterozygous PGC1a mice results in reduced mitochondrial response to changes in ΔGATP and tissue ATP following fasting. These impairments in liver energy state are associated with larger and longer meals during chow feeding, impaired dose-dependent food intake inhibition in response to mixed and individual nutrient oral pre-loads, and greater acute fasting-induced food intake. These data support previous work proposing liver-mediated food intake regulation through modulation of peripheral satiation signals.
Collapse
Affiliation(s)
- Michael E. Ponte
- Dept. of Cell Biology & Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - John C. Prom
- Dept. of Cell Biology & Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Mallory A. Newcomb
- Dept. of Cell Biology & Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Annabelle B. Jordan
- Dept. of Cell Biology & Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Lucas L. Comfort
- Dept. of Cell Biology & Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Jiayin Hu
- Dept. of Psychiatry, University of Pennsylvania, Philadelphia, PA
| | - Patrycja Puchalska
- Division of Molecular Medicine, University of Minnesota, Minneapolis, MN
| | - Caroline E. Geisler
- Dept. of Psychiatry, University of Pennsylvania, Philadelphia, PA
- Dept. of Pharmaceutical Sciences, University of Kentucky, Lexington, KY
| | - Matthew R. Hayes
- Dept. of Psychiatry, University of Pennsylvania, Philadelphia, PA
| | - E. Matthew Morris
- Dept. of Cell Biology & Physiology, University of Kansas Medical Center, Kansas City, Kansas
- Center for Children’s Healthy Lifestyle and Nutrition, Children’s Mercy Hospital, Kansas City, Missouri
- University of Kansas Diabetes Institute, Kansas City, Kansas
| |
Collapse
|
2
|
Sakamoto S, Kakehi S, Abudurezake A, Kaga H, Someya Y, Tabata H, Yoshizawa Y, Naito H, Tajima T, Ito N, Kawamori R, Watada H, Tamura Y. Sex-specific impact of GCKR rs1260326 polymorphism on metabolic traits in an older Japanese population: the Bunkyo Health Study. Ther Adv Endocrinol Metab 2024; 15:20420188241280540. [PMID: 39346030 PMCID: PMC11437585 DOI: 10.1177/20420188241280540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/19/2024] [Indexed: 10/01/2024] Open
Abstract
Background Metabolic syndrome involves health problems influenced by aging and genetics. The glucokinase regulatory protein (GCKR) rs1260326 polymorphism (Leu446) is associated with metabolic traits. This study explores the impact of the GCKR rs1260326 polymorphism on metabolic traits in older Japanese with focusing on sex-specific differences. Methods This cross-sectional study from the Bunkyo Health Study in Tokyo, Japan, examined 883 participants aged 65-84 years. Participants were excluded with diabetes, or on drug treatment for diabetes or dyslipidemia. The GCKR P446L polymorphism was analyzed and compared their characteristics of physical activity, dietary intake, body composition, and metabolic parameters. Results Study participants with GCKR rs1260326 genotypes (C/C 20.7%, C/T 47.6%, T/T 31.7%) had a median age of 72 years, and 60.4% were women. Men with the T/T genotype, as compared to the C/C genotype, had a lower body weight, body mass index (BMI), and skeletal mass index. This genotype also associated with lower fasting insulin, homeostasis model assessment of insulin resistance index (HOMA-IR), and higher Matsuda index, but not after adjustment for age, BMI, and physical activity. In contrast, women with the T/T genotype, compared to the C/C genotype, showed higher C-reactive protein, fibroblast growth factor 21, and Matsuda index. They also had lower fasting insulin, insulin area under the curve, and HOMA-IR; with these associations being independent of age, BMI, and physical activity. Conclusion The GCKR rs1260326 genotype-affected metabolic traits differentially by sex in older Japanese. This highlights the need to consider sex differences in GCKR-related metabolic outcomes.
Collapse
Affiliation(s)
- Shota Sakamoto
- Department of Sports Medicine and Sportology, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Saori Kakehi
- Sportology Center, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Sports Medicine and Sportology, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Abulaiti Abudurezake
- Sportology Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Hideyoshi Kaga
- Metabolism and Endocrinology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Yuki Someya
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
| | - Hiroki Tabata
- Sportology Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Yasuyo Yoshizawa
- Center for Healthy Life Expectancy, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Hitoshi Naito
- Metabolism and Endocrinology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Tsubasa Tajima
- Metabolism and Endocrinology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Naoaki Ito
- Metabolism and Endocrinology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Ryuzo Kawamori
- Department of Sports Medicine and Sportology, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
- Sportology Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Metabolism and Endocrinology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Hirotaka Watada
- Sportology Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Metabolism and Endocrinology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Yoshifumi Tamura
- Department of Sports Medicine and Sportology, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
- Sportology Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Metabolism and Endocrinology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Center for Healthy Life Expectancy, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
- Faculty of International Liberal Arts, Juntendo University, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
3
|
Malla A, Gupta S, Sur R. Glycolytic enzymes in non-glycolytic web: functional analysis of the key players. Cell Biochem Biophys 2024; 82:351-378. [PMID: 38196050 DOI: 10.1007/s12013-023-01213-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/26/2023] [Indexed: 01/11/2024]
Abstract
To survive in the tumour microenvironment, cancer cells undergo rapid metabolic reprograming and adaptability. One of the key characteristics of cancer is increased glycolytic selectivity and decreased oxidative phosphorylation (OXPHOS). Apart from ATP synthesis, glycolysis is also responsible for NADH regeneration and macromolecular biosynthesis, such as amino acid biosynthesis and nucleotide biosynthesis. This allows cancer cells to survive and proliferate even in low-nutrient and oxygen conditions, making glycolytic enzymes a promising target for various anti-cancer agents. Oncogenic activation is also caused by the uncontrolled production and activity of glycolytic enzymes. Nevertheless, in addition to conventional glycolytic processes, some glycolytic enzymes are involved in non-canonical functions such as transcriptional regulation, autophagy, epigenetic changes, inflammation, various signaling cascades, redox regulation, oxidative stress, obesity and fatty acid metabolism, diabetes and neurodegenerative disorders, and hypoxia. The mechanisms underlying the non-canonical glycolytic enzyme activities are still not comprehensive. This review summarizes the current findings on the mechanisms fundamental to the non-glycolytic actions of glycolytic enzymes and their intermediates in maintaining the tumor microenvironment.
Collapse
Affiliation(s)
- Avirup Malla
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, Kolkata, India
| | - Suvroma Gupta
- Department of Aquaculture Management, Khejuri college, West Bengal, Baratala, India.
| | - Runa Sur
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, Kolkata, India.
| |
Collapse
|
4
|
Abuelazm MT, Mohamed I, Naeem A, Khlidj Y, Tanashat M, Katamesh BE, Abusuilik H, Altobaishat O, Abdelnabi M, Abdelazeem B. Intermittent fasting regimens for metabolic dysfunction-associated steatotic liver disease: a systematic review and network meta-analysis of randomized controlled trials. Eur J Gastroenterol Hepatol 2024; 36:371-381. [PMID: 38407890 DOI: 10.1097/meg.0000000000002715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent metabolic disorder characterized by excessive hepatic fat accumulation. Intermittent fasting (IF) has emerged as a potential therapeutic strategy with the ability to induce weight loss, improve insulin sensitivity and reduce hepatic steatosis. We aim to compare the efficacy of different IF regimens for MASLD management. A systematic review and network meta-analysis of randomized controlled trials investigating different IF regimens for MASLD. PubMed , EMBASE , WOS , SCOPUS and Cochrane Central Register of Controlled Trials were searched until 10 April 2023. Analysis was performed using R software with the meta and netmeta packages. Mean difference (MD) was used to pool continuous outcomes with 95% confidence intervals (CIs). Our meta-analysis was registered in PROSPERO (CRD42023418467). Our meta-analysis included eight randomized controlled trials with a total of 635 participants. The 5 : 2 diet significantly improved liver stiffness (MD, -0.32; 95% CI, -0.55 to -0.09; P < 0.01). Time-restricted feeding significantly improved liver steatosis (controlled attenuation parameter score) (MD, -39.83; 95% CI, -64.78 to -14.87; P < 0.01). No significant changes were observed in asparate aminotransferase, gamma-glutamyl transpeptidase, low-density lipoproteins cholesterol, total cholesterol, triglyceride levels, basal metabolic index, blood pressure, Homeostatic Model Assessment of Insulin Resistance, fasting blood sugar, lean body mass or waist circumference across all IF regimens. However, alternate-day fasting showed positive results in anthropometric measures, including significant improvements in lean body mass, waist circumference, fat mass and weight reduction ( P < 0.05). IF regimens showed various positive effects on clinical outcomes in MASLD patients; however, these effects were not consistent. Therefore, a patient-tailored IF regimen should be considered.
Collapse
Affiliation(s)
| | - Islam Mohamed
- Department of Internal Medicine, University of Missouri, Kansas, Missouri, USA
| | - Ahmed Naeem
- Faculty of Medicine, Al-Azhar University, Asyut, Egypt
| | - Yehya Khlidj
- Faculty of Medicine, University of Algiers, Algiers, Algeria
| | | | | | | | - Obieda Altobaishat
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Mohamed Abdelnabi
- Department of Clinical Pharmacy, University of Michigan, Ann Arbor, Michigan, USA
| | - Basel Abdelazeem
- Department of Cardiology, West Virginia University, West Virginia, USA
| |
Collapse
|
5
|
Wang S, Zhang J, Li J, Wang J, Liu W, Zhang Z, Yu H. Label-free quantitative proteomics reveals the potential mechanisms of insoluble dietary fiber from okara in improving hepatic lipid metabolism of high-fat diet-induced mice. J Proteomics 2023; 287:104980. [PMID: 37499746 DOI: 10.1016/j.jprot.2023.104980] [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: 05/17/2022] [Revised: 03/21/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
The high purity insoluble dietary fiber (IDF) from okara is a natural component with a potentially positive effect on a high-fat diet (HFD)-induced hepatic metabolic disorders, although its regulatory mechanism remains unclear. This study aims to elucidate the potential pathways and key proteins of IDF for the amelioration of hepatic lipid metabolism in mice fed with HFD. Here, we used label-free quantitative proteomics technology to quantity and identify differentially expressed proteins in the liver that are associated with IDF treatment. The differentially expressed proteins were assessed by GO annotation and KEGG pathways. Western blot and qRT-PCR analyses were conducted to validate the potential targets regulated by IDF. In total, 73 differentially expressed proteins were identified, of which 27 were up-regulated (FC > 1.5) and 46 were down-regulated (FC < 0.667). GO analysis suggested that differentially expressed proteins were mainly located in the cell and organelles, regulated biological processes, and were associated with enzyme activity and molecular binding. The KEGG pathway enrichment analysis further demonstrated glycolysis/gluconeogenesis, pyruvate metabolism, TCA cycle, arginine biosynthesis, alanine, aspartate and glutamate metabolism, and retinol metabolism were affected. The combination of proteomics, Western blot, and qRT-PCR suggested that ACS, ACLY, GOT1, GLS2, NAGS, CYP4A10, CYP3A25, and CYP2A5 in these pathways might be key proteins for IDF intervention. Taken together, our findings elucidate new mechanisms involved in how IDF affects hepatic metabolism, provide important information for the functional food industries, and improve the added value of okara. SIGNIFICANCE: Okara is evidenced as a high-quality by-product with several nutritional components, especially dietary fiber (50-60%) labeled as "The Seventh Nutrient". Previous studies have shown that IDF has a positive potential effect on a high-fat diet (HFD)-induced hepatic metabolic disorders, but its molecular mechanism remains unclear. To elucidate the therapeutic mechanism of IDF at the protein level, a label-free quantitative proteomic analysis was used to identify the dynamic changes of the liver proteome between HIDF and HFD groups in this study. These results provide a new perspective for exploring the therapeutic mechanism of IDF at the protein level and enlightenment for promoting the comprehensive utilization of okara.
Collapse
Affiliation(s)
- Sainan Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, China; Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun, Jilin 130118, China
| | - Jiarui Zhang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, China; Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun, Jilin 130118, China
| | - Jiaxin Li
- Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun, Jilin 130118, China; Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical and Food Chemistry, Faculty of Sciences, Ourense, 32004, Spain
| | - Junyao Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, China; Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun, Jilin 130118, China
| | - Wenhao Liu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, China; Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun, Jilin 130118, China
| | - Zhao Zhang
- Shandong Sinoglory Health Food Co., Ltd., Liaocheng, Shandong 252000, China
| | - Hansong Yu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, China; Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun, Jilin 130118, China.
| |
Collapse
|
6
|
Naomi R, Teoh SH, Halim S, Embong H, Hasain Z, Bahari H, Kumar J. Unraveling Obesity: Transgenerational Inheritance, Treatment Side Effects, Flavonoids, Mechanisms, Microbiota, Redox Balance, and Bioavailability-A Narrative Review. Antioxidants (Basel) 2023; 12:1549. [PMID: 37627544 PMCID: PMC10451614 DOI: 10.3390/antiox12081549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/02/2023] [Accepted: 07/06/2023] [Indexed: 08/27/2023] Open
Abstract
Obesity is known as a transgenerational vicious cycle and has become a global burden due to its unavoidable complications. Modern approaches to obesity management often involve the use of pharmaceutical drugs and surgeries that have been associated with negative side effects. In contrast, natural antioxidants, such as flavonoids, have emerged as a promising alternative due to their potential health benefits and minimal side effects. Thus, this narrative review explores the potential protective role of flavonoids as a natural antioxidant in managing obesity. To identify recent in vivo studies on the efficiency of flavonoids in managing obesity, a comprehensive search was conducted on Wiley Online Library, Scopus, Nature, and ScienceDirect. The search was limited to the past 10 years; from the search, we identified 31 articles to be further reviewed. Based on the reviewed articles, we concluded that flavonoids offer novel therapeutic strategies for preventing obesity and its associated co-morbidities. This is because the appropriate dosage of flavonoid compounds is able to reduce adipose tissue mass, the formation of intracellular free radicals, enhance endogenous antioxidant defences, modulate the redox balance, and reduce inflammatory signalling pathways. Thus, this review provides an insight into the domain of a natural product therapeutic approach for managing obesity and recapitulates the transgenerational inheritance of obesity, the current available treatments to manage obesity and its side effects, flavonoids and their sources, the molecular mechanism involved, the modulation of gut microbiota in obesity, redox balance, and the bioavailability of flavonoids. In toto, although flavonoids show promising positive outcome in managing obesity, a more comprehensive understanding of the molecular mechanisms responsible for the advantageous impacts of flavonoids-achieved through translation to clinical trials-would provide a novel approach to inculcating flavonoids in managing obesity in the future as this review is limited to animal studies.
Collapse
Affiliation(s)
- Ruth Naomi
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia;
| | - Soo Huat Teoh
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas 13200, Malaysia;
| | - Shariff Halim
- Faculty of Health Sciences, University Technology Mara (UiTM) Pulau Pinang, Bertam Campus, Kepala Batas 13200, Malaysia;
| | - Hashim Embong
- Department of Emergency Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Zubaidah Hasain
- Unit of Physiology, Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia, Kuala Lumpur 57000, Malaysia
| | - Hasnah Bahari
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia;
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
7
|
Ramatchandirin B, Pearah A, He L. Regulation of Liver Glucose and Lipid Metabolism by Transcriptional Factors and Coactivators. Life (Basel) 2023; 13:life13020515. [PMID: 36836874 PMCID: PMC9962321 DOI: 10.3390/life13020515] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
The prevalence of nonalcoholic fatty liver disease (NAFLD) worldwide is on the rise and NAFLD is becoming the most common cause of chronic liver disease. In the USA, NAFLD affects over 30% of the population, with similar occurrence rates reported from Europe and Asia. This is due to the global increase in obesity and type 2 diabetes mellitus (T2DM) because patients with obesity and T2DM commonly have NAFLD, and patients with NAFLD are often obese and have T2DM with insulin resistance and dyslipidemia as well as hypertriglyceridemia. Excessive accumulation of triglycerides is a hallmark of NAFLD and NAFLD is now recognized as the liver disease component of metabolic syndrome. Liver glucose and lipid metabolisms are intertwined and carbon flux can be used to generate glucose or lipids; therefore, in this review we discuss the important transcription factors and coactivators that regulate glucose and lipid metabolism.
Collapse
Affiliation(s)
| | - Alexia Pearah
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ling He
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Baltimore, MD 21287, USA
- Correspondence: ; Tel.: +1-410-502-5765; Fax: +1-410-502-5779
| |
Collapse
|
8
|
Narrative Review: Glucocorticoids in Alcoholic Hepatitis—Benefits, Side Effects, and Mechanisms. J Xenobiot 2022; 12:266-288. [PMID: 36278756 PMCID: PMC9589945 DOI: 10.3390/jox12040019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/03/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
Alcoholic hepatitis is a major health and economic burden worldwide. Glucocorticoids (GCs) are the only first-line drugs recommended to treat severe alcoholic hepatitis (sAH), with limited short-term efficacy and significant side effects. In this review, I summarize the major benefits and side effects of GC therapy in sAH and the potential underlying mechanisms. The review of the literature and data mining clearly indicate that the hepatic signaling of glucocorticoid receptor (GR) is markedly impaired in sAH patients. The impaired GR signaling causes hepatic down-regulation of genes essential for gluconeogenesis, lipid catabolism, cytoprotection, and anti-inflammation in sAH patients. The efficacy of GCs in sAH may be compromised by GC resistance and/or GC’s extrahepatic side effects, particularly the side effects of intestinal epithelial GR on gut permeability and inflammation in AH. Prednisolone, a major GC used for sAH, activates both the GR and mineralocorticoid receptor (MR). When GC non-responsiveness occurs in sAH patients, the activation of MR by prednisolone might increase the risk of alcohol abuse, liver fibrosis, and acute kidney injury. To improve the GC therapy of sAH, the effort should be focused on developing the biomarker(s) for GC responsiveness, liver-targeting GR agonists, and strategies to overcome GC non-responsiveness and prevent alcohol relapse in sAH patients.
Collapse
|
9
|
Zhang F, Zuo T, Wan Y, Xu Z, Cheung C, Li AY, Zhu W, Tang W, Chan PK, Chan FK, Ng SC. Multi-omic analyses identify mucosa bacteria and fecal metabolites associated with weight loss after fecal microbiota transplantation. Innovation (N Y) 2022; 3:100304. [PMID: 36091491 PMCID: PMC9460156 DOI: 10.1016/j.xinn.2022.100304] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 08/13/2022] [Indexed: 11/19/2022] Open
Abstract
Fecal microbiota transplantation (FMT) has shown promising results in animal models of obesity, while results in human studies are inconsistent. We aimed to determine factors associated with weight loss after FMT in nine obese subjects using serial multi-omics analysis of the fecal and mucosal microbiome. The mucosal microbiome, fecal microbiome, and fecal metabolome showed individual clustering in each subject after FMT. The colonic microbiome in patients showed more marked variance after FMT compared with the duodenal microbiome, characterized by an increased relative abundance of Bacteroides. Subjects who lost weight after FMT sustained enrichment of Bifidobacterium bifidum and Alistipes onderdonkii in the duodenal, colonic mucosal, and fecal microbiome and increased levels of phosphopantothenate biosynthesis and fecal metabolite eicosapentaenoic acid (EPA), compared with those without weight loss. Fecal levels of amino acid metabolism-associated were positively correlated with the fecal abundance of B. bifidum, and fatty acid metabolism-associated metabolites showed positive correlations with A. onderdonkii. We report for the first time the individualized response of fecal and mucosa microbiome to FMT in obese subjects and highlight that FMT is less capable of shaping the small intestine microbiota. These findings contribute to personalized microbe-based therapies for obesity.
Collapse
Affiliation(s)
- Fen Zhang
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Microbiota I-Center (MagIC), Hong Kong 999077, China
| | - Tao Zuo
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Microbiota I-Center (MagIC), Hong Kong 999077, China
| | - Yating Wan
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Microbiota I-Center (MagIC), Hong Kong 999077, China
| | - Zhilu Xu
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Microbiota I-Center (MagIC), Hong Kong 999077, China
| | - Chunpan Cheung
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Microbiota I-Center (MagIC), Hong Kong 999077, China
| | - Amy Y. Li
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Wenyi Zhu
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Microbiota I-Center (MagIC), Hong Kong 999077, China
| | - Whitney Tang
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Microbiota I-Center (MagIC), Hong Kong 999077, China
| | - Paul K.S. Chan
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong 999077, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Francis K.L. Chan
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Microbiota I-Center (MagIC), Hong Kong 999077, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Siew C. Ng
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Microbiota I-Center (MagIC), Hong Kong 999077, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
- Corresponding author
| |
Collapse
|
10
|
Mohd Ghozali N, Giribabu N, Salleh N. Mechanisms Linking Vitamin D Deficiency to Impaired Metabolism: An Overview. Int J Endocrinol 2022; 2022:6453882. [PMID: 35859985 PMCID: PMC9293580 DOI: 10.1155/2022/6453882] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/19/2022] [Accepted: 06/13/2022] [Indexed: 12/12/2022] Open
Abstract
Vitamin D deficiency is a common health problem worldwide. Despite its known skeletal effects, studies have begun to explore its extra-skeletal effects, that is, in preventing metabolic diseases such as obesity, hyperlipidemia, and diabetes mellitus. The mechanisms by which vitamin D deficiency led to these unfavorable metabolic consequences have been explored. Current evidence indicates that the deficiency of vitamin D could impair the pancreatic β-cell functions, thus compromising its insulin secretion. Besides, vitamin D deficiency could also exacerbate inflammation, oxidative stress, and apoptosis in the pancreas and many organs, which leads to insulin resistance. Together, these will contribute to impairment in glucose homeostasis. This review summarizes the reported metabolic effects of vitamin D, in order to identify its potential use to prevent and overcome metabolic diseases.
Collapse
Affiliation(s)
- Nurulmuna Mohd Ghozali
- Department of Physiology, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur 59100, Malaysia
| | - Nelli Giribabu
- Department of Physiology, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur 59100, Malaysia
| | - Naguib Salleh
- Department of Physiology, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur 59100, Malaysia
| |
Collapse
|
11
|
Li Z, Tang Y, Liu Z, Fan Q, Chen M, Lin Z, Zhao C, Lin R. Hepatotoxicity induced by PPⅥ and PPⅦ in zebrafish were related to the Cholesterol disorder. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 95:153787. [PMID: 34782205 DOI: 10.1016/j.phymed.2021.153787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/15/2021] [Accepted: 09/29/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND PPⅥ2 and PPⅦ3 were a group of Pennogenin compounds extracted from the Paris polyphylla and caused hepatotoxicity in human, while the potential underlying mechanism was unclear. PURPOSE To evaluated the adverse effects of PPⅥ and PPⅦ on the liver in the zebrafish. METHOD In this study, 4dpf zebrafish were used for acute toxicity test, LC0 was calculated, and 1/2LC0 and 3/5LC0 were selected for pathological section and liver area measurement to verify the hepatotoxicity of PPⅥ and PPⅦ. Etabonomics study was then conducted to further explore the mechanism of hepatotoxicity of PPⅥ and PPⅦ. Lovastatin was used as an inhibitor, and PCR was used to verify the results. RESULT The result showed that under the condition of sub-lethal concentration exposure, hepatotoxicity-included changes in liver phenotype (liver area), hepatocyte swelling and degeneration, liver cell apoptosis and disturbed biochemical index were observed in zebrafish treated with PPⅥ and PPⅦ. Furthermore, the transcriptome was conducted to confirm the toxicity mechanism shared with PPⅥ and PPⅦ, and we found that steroid biosynthesis process and the related target genes were mainly affected. While, lovastatin treatment effectively ameliorated PPⅦ-induced zebrafish liver injury by improving the liver tissue structure and regulate the expression of associated genes including HMGCRA, SREBP, LSS, CYP2R1, PIK3R3A, GDPD1 and PFKFB-2. CONCLUSION This study was the first investigation to provide the direct evidence of hepatotoxicity of PPⅥ and PPⅦ in vivo zebrafish model, which were related to the steroid biosynthesis. furthermore, in lovastatin played an important role in protection against hepatotoxicity induced by PPVI and PPⅦ by regulating the cholesterol metabolism.
Collapse
Affiliation(s)
- Zhiqi Li
- Beijing University of Chinese Medicine, No. 11 Beisanhuan East Road, Chaoyang, Beijing 100102, China; Beijing Key Lab for Quality Evaluation of Chinese Materia Medica, No. 11 Beisanhuan East Road, Chaoyang, Beijing 100102, China
| | - Yang Tang
- Beijing University of Chinese Medicine, No. 11 Beisanhuan East Road, Chaoyang, Beijing 100102, China
| | - Zhenquan Liu
- Beijing University of Chinese Medicine, No. 11 Beisanhuan East Road, Chaoyang, Beijing 100102, China
| | - Qiqi Fan
- Beijing University of Chinese Medicine, No. 11 Beisanhuan East Road, Chaoyang, Beijing 100102, China; Beijing Key Lab for Quality Evaluation of Chinese Materia Medica, No. 11 Beisanhuan East Road, Chaoyang, Beijing 100102, China
| | - Meilin Chen
- Beijing University of Chinese Medicine, No. 11 Beisanhuan East Road, Chaoyang, Beijing 100102, China; Beijing Key Lab for Quality Evaluation of Chinese Materia Medica, No. 11 Beisanhuan East Road, Chaoyang, Beijing 100102, China
| | - Zili Lin
- Beijing University of Chinese Medicine, No. 11 Beisanhuan East Road, Chaoyang, Beijing 100102, China
| | - Chongjun Zhao
- Beijing University of Chinese Medicine, No. 11 Beisanhuan East Road, Chaoyang, Beijing 100102, China; Beijing Key Lab for Quality Evaluation of Chinese Materia Medica, No. 11 Beisanhuan East Road, Chaoyang, Beijing 100102, China.
| | - Ruichao Lin
- Beijing University of Chinese Medicine, No. 11 Beisanhuan East Road, Chaoyang, Beijing 100102, China; Beijing Key Lab for Quality Evaluation of Chinese Materia Medica, No. 11 Beisanhuan East Road, Chaoyang, Beijing 100102, China.
| |
Collapse
|
12
|
Recent Advances in Adipose Tissue Dysfunction and Its Role in the Pathogenesis of Non-Alcoholic Fatty Liver Disease. Cells 2021; 10:cells10123300. [PMID: 34943809 PMCID: PMC8699427 DOI: 10.3390/cells10123300] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/09/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity is a serious ongoing health problem that significantly increases the incidence of nonalcoholic fatty liver disease (NAFLD). During obesity, adipose tissue dysfunction is obvious and characterized by increased fat deposition (adiposity) and chronic low-grade inflammation. The latter has been implicated to critically promote the development and progression of NAFLD, whose advanced form non-alcoholic steatohepatitis (NASH) is considered one of the most common causes of terminal liver diseases. This review summarizes the current knowledge on obesity-related adipose dysfunction and its roles in the pathogenesis of hepatic steatosis and inflammation, as well as liver fibrosis. A better understanding of the crosstalk between adipose tissue and liver under obesity is essential for the development of new and improved preventive and/or therapeutic approaches for managing NAFLD.
Collapse
|
13
|
Genome-wide discovery of hidden genes mediating known drug-disease association using KDDANet. NPJ Genom Med 2021; 6:50. [PMID: 34131148 PMCID: PMC8206141 DOI: 10.1038/s41525-021-00216-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 05/25/2021] [Indexed: 11/09/2022] Open
Abstract
Many of genes mediating Known Drug-Disease Association (KDDA) are escaped from experimental detection. Identifying of these genes (hidden genes) is of great significance for understanding disease pathogenesis and guiding drug repurposing. Here, we presented a novel computational tool, called KDDANet, for systematic and accurate uncovering the hidden genes mediating KDDA from the perspective of genome-wide functional gene interaction network. KDDANet demonstrated the competitive performances in both sensitivity and specificity of identifying genes in mediating KDDA in comparison to the existing state-of-the-art methods. Case studies on Alzheimer's disease (AD) and obesity uncovered the mechanistic relevance of KDDANet predictions. Furthermore, when applied with multiple types of cancer-omics datasets, KDDANet not only recapitulated known genes mediating KDDAs related to cancer, but also revealed novel candidates that offer new biological insights. Importantly, KDDANet can be used to discover the shared genes mediating multiple KDDAs. KDDANet can be accessed at http://www.kddanet.cn and the code can be freely downloaded at https://github.com/huayu1111/KDDANet .
Collapse
|
14
|
Zhu B, Guo X, Xu H, Jiang B, Li H, Wang Y, Yin Q, Zhou T, Cai JJ, Glaser S, Meng F, Francis H, Alpini G, Wu C. Adipose tissue inflammation and systemic insulin resistance in mice with diet-induced obesity is possibly associated with disruption of PFKFB3 in hematopoietic cells. J Transl Med 2021; 101:328-340. [PMID: 33462362 PMCID: PMC7897240 DOI: 10.1038/s41374-020-00523-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/13/2020] [Accepted: 11/28/2020] [Indexed: 02/06/2023] Open
Abstract
Obesity-associated inflammation in white adipose tissue (WAT) is a causal factor of systemic insulin resistance; however, precisely how immune cells regulate WAT inflammation in relation to systemic insulin resistance remains to be elucidated. The present study examined a role for 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) in hematopoietic cells in regulating WAT inflammation and systemic insulin sensitivity. Male C57BL/6J mice were fed a high-fat diet (HFD) or low-fat diet (LFD) for 12 weeks and examined for WAT inducible 6-phosphofructo-2-kinase (iPFK2) content, while additional HFD-fed mice were treated with rosiglitazone and examined for PFKFB3 mRNAs in WAT stromal vascular cells (SVC). Also, chimeric mice in which PFKFB3 was disrupted only in hematopoietic cells and control chimeric mice were also fed an HFD and examined for HFD-induced WAT inflammation and systemic insulin resistance. In vitro, adipocytes were co-cultured with bone marrow-derived macrophages and examined for adipocyte proinflammatory responses and insulin signaling. Compared with their respective levels in controls, WAT iPFK2 amount in HFD-fed mice and WAT SVC PFKFB3 mRNAs in rosiglitazone-treated mice were significantly increased. When the inflammatory responses were analyzed, peritoneal macrophages from PFKFB3-disrputed mice revealed increased proinflammatory activation and decreased anti-inflammatory activation compared with control macrophages. At the whole animal level, hematopoietic cell-specific PFKFB3 disruption enhanced the effects of HFD feeding on promoting WAT inflammation, impairing WAT insulin signaling, and increasing systemic insulin resistance. In vitro, adipocytes co-cultured with PFKFB3-disrupted macrophages revealed increased proinflammatory responses and decreased insulin signaling compared with adipocytes co-cultured with control macrophages. These results suggest that PFKFB3 disruption in hematopoietic cells only exacerbates HFD-induced WAT inflammation and systemic insulin resistance.
Collapse
Affiliation(s)
- Bilian Zhu
- Department of Nutrition, Texas A&M University, College Station, TX, USA
- Department of VIP Medical Service Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xin Guo
- Department of Nutrition, Texas A&M University, College Station, TX, USA
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Shandong, Jinan, China
| | - Hang Xu
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Boxiong Jiang
- Department of VIP Medical Service Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Honggui Li
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Yina Wang
- Department of VIP Medical Service Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qiongli Yin
- Department of VIP Medical Service Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Tianhao Zhou
- Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, USA
| | - James J Cai
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Shannon Glaser
- Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, USA
| | - Fanyin Meng
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, IN, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Heather Francis
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, IN, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Gianfranco Alpini
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, IN, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Chaodong Wu
- Department of Nutrition, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
15
|
Bordeleau M, Fernández de Cossío L, Chakravarty MM, Tremblay MÈ. From Maternal Diet to Neurodevelopmental Disorders: A Story of Neuroinflammation. Front Cell Neurosci 2021; 14:612705. [PMID: 33536875 PMCID: PMC7849357 DOI: 10.3389/fncel.2020.612705] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
Providing the appropriate quantity and quality of food needed for both the mother's well-being and the healthy development of the offspring is crucial during pregnancy. However, the macro- and micronutrient intake also impacts the body's regulatory supersystems of the mother, such as the immune, endocrine, and nervous systems, which ultimately influence the overall development of the offspring. Of particular importance is the association between unhealthy maternal diet and neurodevelopmental disorders in the offspring. Epidemiological studies have linked neurodevelopmental disorders like autism spectrum disorders, attention-deficit-hyperactivity disorder, and schizophrenia, to maternal immune activation (MIA) during gestation. While the deleterious consequences of diet-induced MIA on offspring neurodevelopment are increasingly revealed, neuroinflammation is emerging as a key underlying mechanism. In this review, we compile the evidence available on how the mother and offspring are both impacted by maternal dietary imbalance. We specifically explore the various inflammatory and anti-inflammatory effects of dietary components and discuss how changes in inflammatory status can prime the offspring brain development toward neurodevelopmental disorders. Lastly, we discuss research evidence on the mechanisms that sustain the relationship between maternal dietary imbalance and offspring brain development, involving altered neuroinflammatory status in the offspring, as well as genetic to cellular programming notably of microglia, and the evidence that the gut microbiome may act as a key mediator.
Collapse
Affiliation(s)
- Maude Bordeleau
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | | | - M. Mallar Chakravarty
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
- Cerebral Imaging Centre, Douglas Mental Health University, McGill University, Montréal, QC, Canada
- Department of Psychiatry, McGill University, Montréal, QC, Canada
- Department of Biological and Biomedical Engineering, McGill University, Montréal, QC, Canada
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Département de Médecine Moléculaire, Université Laval, Québec, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Biochemistry and Molecular Biology, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
16
|
Peng X, Yu Q, Liu Y, Ma T, Li M. Study on the Function of the Inositol Polyphosphate Kinases Kcs1 and Vip1 of Candida albicans in Energy Metabolism. Front Microbiol 2020; 11:566069. [PMID: 33362729 PMCID: PMC7758236 DOI: 10.3389/fmicb.2020.566069] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 11/11/2020] [Indexed: 12/15/2022] Open
Abstract
In Saccharomyces cerevisiae, inositol polyphosphate kinase KCS1 but not VIP1 knockout is of great significance for maintaining cell viability, promoting glycolysis metabolism, and inducing mitochondrial damage. The functions of Candida albicans inositol polyphosphate kinases Kcs1 and Vip1 have not yet been studied. In this study, we found that the growth rate of C. albicans vip1Δ/Δ strain in glucose medium was reduced and the upregulation of glycolysis was accompanied by a decrease in mitochondrial activity, resulting in a large accumulation of lipid droplets, along with an increase in cell wall chitin and cell membrane permeability, eventually leading to cell death. Relieving intracellular glycolysis rate or increasing mitochondrial metabolism can reduce lipid droplet accumulation, causing a reduction in chitin content and cell membrane permeability. The growth activity and energy metabolism of the vip1Δ/Δ strains in a non-fermentable carbon source glycerol medium were not different from those of the wild-type strains, indicating that knocking out VIP1 did not cause mitochondria damage. Moreover, C. albicans KCS1 knockout did not affect cell activity and energy metabolism. Thus, in C. albicans, Vip1 is more important than Kcs1 in regulating cell viability and energy metabolism.
Collapse
Affiliation(s)
- Xueling Peng
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Qilin Yu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Yingzheng Liu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Tianyu Ma
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Mingchun Li
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
17
|
Cao X, Guo Y, Wang Y, Wang H, Liu D, Gong Y, Wang J, Chen X, Zhang W. Effects of high-fat diet and Apoe deficiency on retinal structure and function in mice. Sci Rep 2020; 10:18601. [PMID: 33139746 PMCID: PMC7606505 DOI: 10.1038/s41598-020-75576-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 10/16/2020] [Indexed: 12/17/2022] Open
Abstract
To investigate the effects of a high-fat diet (HFD) and apolipoprotein E (Apoe) deficiency on retinal structure and function in mice. Apoe KO mice and wild-type C57BL/6J mice were given a low-fat diet (LFD) or a HFD for 32 weeks. Blood glucose, serum lipids, body weight and visceral fat weight were evaluated. Retinal sterol quantification was carried out by isotope dilution gas chromatography-mass spectrometry. The cholesterol metabolism related genes SCAP-SREBP expressions were detected by qRT-PCR. Retinal function was recorded using an electroretinogram. The thickness of each layer of the retina was measured by optical coherence tomography. Fundus fluorescein angiography was performed to detect retinal vasculature changes. Immunohistochemical staining was used to determine the expression of NF-κB, TNF-α and VEGFR2 in the retina among HFD, HFD Apoe-/-, LFD Apoe-/- and WT mice retinas. HFD feeding caused the mice to gain weight and develop hypercholesterinemia, while Apoe-/- abnormalities also affected blood lipid metabolism. Both HFD and Apoe deficiency elevated retinal cholesterol, especially in the HFD Apoe-/- mice. No up-regulated expression of SCAP-SREBP was observed as a negative regulator. Impaired retinal functions, thinning retinas and abnormal retinal vasculature were observed in the peripheral retinas of the HFD and Apoe-/- mice compared with those in the normal chow group, particularly in the HFD Apoe-/- mice. Moreover, the expression of NF-κB in the retinas of the HFD and Apoe-/- mice was increased, together with upregulated TNF-α mRNA levels and TNF-α expression in the layer of retinal ganglion cells of the peripheral retina. At the same time, the expression level of VEGFR2 was elevated in the intervention groups, most notably in HFD Apoe-/- mice. HFD or Apoe gene deletion had certain adverse effects on retinal function and structure, which were far below the combined factors and induced harm to the retina. Furthermore, HFD caused retinal ischemia and hypoxia. Additionally, Apoe abnormality increased susceptibility to ischemia. These changes upregulated NF-κB expression in ganglion cells and activated downstream TNF-α. Simultaneously, they activated VEGFR2, accelerating angiogenesis and vascular permeability. All of the aforementioned outcomes initiated inflammatory responses to trigger ganglion cell apoptosis and aggravate retinal neovascularization.
Collapse
Affiliation(s)
- Xiupeng Cao
- Tianjin Medical University, Tianjin, China.,Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin, China.,Nankai University Affiliated Eye Hospital, Tianjin, China
| | - Yatu Guo
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin, China. .,Nankai University Affiliated Eye Hospital, Tianjin, China.
| | - Yuchuan Wang
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin, China.,Nankai University Affiliated Eye Hospital, Tianjin, China
| | - Hao Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin, China
| | - Dong Liu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin, China
| | - Yibo Gong
- Tianjin Medical University, Tianjin, China.,Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin, China.,Nankai University Affiliated Eye Hospital, Tianjin, China
| | - Jue Wang
- Tianjin Medical University, Tianjin, China.,Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin, China.,Nankai University Affiliated Eye Hospital, Tianjin, China
| | - Xia Chen
- Tianjin Medical University, Tianjin, China. .,Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin, China. .,Nankai University Affiliated Eye Hospital, Tianjin, China.
| | - Wei Zhang
- Tianjin Medical University, Tianjin, China. .,Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin, China. .,Nankai University Affiliated Eye Hospital, Tianjin, China.
| |
Collapse
|
18
|
Li R, Huang X, Liang X, Su M, Lai KP, Chen J. Integrated omics analysis reveals the alteration of gut microbe-metabolites in obese adults. Brief Bioinform 2020; 22:5882185. [PMID: 32770198 DOI: 10.1093/bib/bbaa165] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022] Open
Abstract
Obesity, a risk to health, is a global problem in modern society. The prevalence of obesity was approximately 13% among world's adult population. Recently, several reports suggested that the interference of gut microbiota composition and function is associated with metabolic disorders, including obesity. Gut microbiota produce a board range of metabolites involved in energy and glucose homeostasis, leading to the alteration in host metabolism. However, systematic evaluation of the relationship between gut microbiota, gut metabolite and host metabolite profiles in obese adults is still lacking. In this study, we used comparative metagenomics and metabolomics analysis to determine the gut microbiota and gut-host metabolite profiles in six normal and obese adults of Chinese origin, respectively. Following the functional and pathway analysis, we aimed to understand the possible impact of gut microbiota on the host metabolites via the change in gut metabolites. The result showed that the change in gut microbiota may result in the modulation of gut metabolites contributing to glycolysis, tricarboxylic acid cycle and homolactic fermentation. Furthermore, integrated metabolomic analysis demonstrated a possible positive correlation of dysregulated metabolites in the gut and host, including l-phenylalanine, l-tyrosine, uric acid, kynurenic acid, cholesterol sulfate and glucosamine, which were reported to contribute to metabolic disorders such as obesity and diabetes. The findings of this study provide the possible association between gut microbiota-metabolites and host metabolism in obese adults. The identified metabolite changes could serve as biomarkers for the evaluation of obesity and metabolic disorders.
Collapse
Affiliation(s)
| | | | | | - Min Su
- Guilin Medical University
| | | | | |
Collapse
|
19
|
Kleinert M, Sachs S, Habegger KM, Hofmann SM, Müller TD. Glucagon Regulation of Energy Expenditure. Int J Mol Sci 2019; 20:ijms20215407. [PMID: 31671603 PMCID: PMC6862306 DOI: 10.3390/ijms20215407] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 02/07/2023] Open
Abstract
Glucagon's ability to increase energy expenditure has been known for more than 60 years, yet the mechanisms underlining glucagon's thermogenic effect still remain largely elusive. Over the last years, significant efforts were directed to unravel the physiological and cellular underpinnings of how glucagon regulates energy expenditure. In this review, we summarize the current knowledge on how glucagon regulates systems metabolism with a special emphasis on its acute and chronic thermogenic effects.
Collapse
Affiliation(s)
- Maximilian Kleinert
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Centre Munich, Ingolstädter Landstraße 1, 85764 Oberschleißheim, Germany.
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, 2100 Copenhagen, Denmark.
| | - Stephan Sachs
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Centre Munich, Ingolstädter Landstraße 1, 85764 Oberschleißheim, Germany.
- Division of Metabolic Diseases, Technische Universität München, 85740 Munich, Germany.
| | - Kirk M Habegger
- Department of Medicine-Endocrinology and Comprehensive Diabetes Center, Diabetes and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35899, USA.
| | - Susanna M Hofmann
- Institute for Diabetes and Regeneration, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany.
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany.
- Medizinische Klinik und Poliklinik IV, Klinikum der LMU, 80336 München, Germany.
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Centre Munich, Ingolstädter Landstraße 1, 85764 Oberschleißheim, Germany.
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany.
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics, 72076 Tübingen, Germany.
| |
Collapse
|
20
|
Xu Q, Jia H, Ma L, Liu G, Xu C, Li Y, Li X, Li X. All-trans retinoic acid inhibits lipopolysaccharide-induced inflammatory responses in bovine adipocytes via TGFβ1/Smad3 signaling pathway. BMC Vet Res 2019; 15:48. [PMID: 30709353 PMCID: PMC6359792 DOI: 10.1186/s12917-019-1791-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 01/21/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Dairy cows with metabolic disorder in peripartal period display high inflammatory levels. Adipose tissue is a major endocrine organ, which is closely related to systemic inflammation. Retinoic acid (RA), an active metabolite of vitamin A, has shown potential therapeutic immunomodulatory properties. The objective of the study was to examine the effect of all-trans-RA (ATRA), the biologically most active metabolite of vitamin A, on lipopolysaccharide (LPS)-induced bovine adipocytes inflammatory responses and elucidate the underlying mechanisms. Primary cultured bovine adipocytes were treated with ATRA in the presence or absence of LPS. The treated cells were examined for the inflammatory responses and the activity of transforming growth factor beta 1 (TGFβ1) /Smad3 signaling pathway. RESULTS LPS treatment significantly decreased the expression levels of TGFβ1/Smad3 components and increased the content of pro-inflammatory cytokines. Treatment with ATRA could over-activate TGFβ1/Smad3 signaling pathway in bovine adipocytes and reversed the over-production of pro-inflammatory cytokines and inhibition of anti-inflammatory cytokines induced by LPS. Importantly, inhibition of TGFβ1/Smad3 signaling diminished the effects of ATRA on suppressing the proinflammatory responses induced by LPS. Furthermore, activation of TGFβ1/Smad3 signaling further extended the effects of ATRA on suppressing the proinflammatory responses on LPS stimulation. CONCLUSION In conclusion, ATRA stimulates TGFβ1/Smad3 signaling pathway and further suppresses bovine adipocytes inflammatory responses induced by LPS.
Collapse
Affiliation(s)
- Qiushi Xu
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Hongdou Jia
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Li Ma
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Guowen Liu
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Chuang Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Ying Li
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Xinwei Li
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China.
| | - Xiaobing Li
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
21
|
Calorie restriction and its impact on gut microbial composition and global metabolism. Front Med 2018; 12:634-644. [PMID: 30446879 DOI: 10.1007/s11684-018-0670-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 09/27/2018] [Indexed: 02/08/2023]
Abstract
Calorie restriction (CR) is a dietary regimen that reduces calorie intake without incurring malnutrition or a reduction in essential nutrients. It has long been recognized as a natural strategy for promoting health, extending longevity, and prevents the development of metabolic and age-related diseases. In the present review, we focus on the general effect of CR on gut microbiota composition and global metabolism. We also propose mechanisms for its beneficial effect. Results showed that probiotic and butyrate-producing microbes increased their relative abundance, whereas proinflammatory strains exhibited suppressed relative abundance following CR. Analyses of the gut microbial and host metabolisms revealed that most host microbial co-metabolites were changed due to CR. Examples of dramatic CR-induced changes in host metabolism included a decrease in the rate of lipid biosynthesis and an increase in the rates of fatty acid catabolism, β-oxidation, glycogenolysis, and gluconeogenesis. The observed phenotypes and the further verification of the direct link between gut microbiota and metabolome may benefit patients that are at risk for developing metabolic disease. Thus, improved gut microbiota composition and metabolome are potential biomarkers for determining the effectiveness of dietary interventions for age-related and metabolic diseases.
Collapse
|
22
|
Cai Y, Li H, Liu M, Pei Y, Zheng J, Zhou J, Luo X, Huang W, Ma L, Yang Q, Guo S, Xiao X, Li Q, Zeng T, Meng F, Francis H, Glaser S, Chen L, Huo Y, Alpini G, Wu C. Disruption of adenosine 2A receptor exacerbates NAFLD through increasing inflammatory responses and SREBP1c activity. Hepatology 2018; 68:48-61. [PMID: 29315766 PMCID: PMC6033664 DOI: 10.1002/hep.29777] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 12/15/2017] [Accepted: 12/29/2018] [Indexed: 01/04/2023]
Abstract
UNLABELLED Adenosine 2A receptor (A2A R) exerts protective roles in endotoxin- and/or ischemia-induced tissue damage. However, the role for A2A R in nonalcoholic fatty liver disease (NAFLD) remains largely unknown. We sought to examine the effects of global and/or myeloid cell-specific A2A R disruption on the aspects of obesity-associated NAFLD and to elucidate the underlying mechanisms. Global and/or myeloid cell-specific A2A R-disrupted mice and control mice were fed a high-fat diet (HFD) to induce NAFLD. In addition, bone marrow-derived macrophages and primary mouse hepatocytes were examined for inflammatory and metabolic responses. Upon feeding an HFD, both global A2A R-disrupted mice and myeloid cell-specific A2A R-defcient mice revealed increased severity of HFD-induced hepatic steatosis and inflammation compared with their respective control mice. In in vitro experiments, A2A R-deficient macrophages exhibited increased proinflammatory responses, and enhanced fat deposition of wild-type primary hepatocytes in macrophage-hepatocyte cocultures. In primary hepatocytes, A2A R deficiency increased the proinflammatory responses and enhanced the effect of palmitate on stimulating fat deposition. Moreover, A2A R deficiency significantly increased the abundance of sterol regulatory element-binding protein 1c (SREBP1c) in livers of fasted mice and in hepatocytes upon nutrient deprivation. In the absence of A2A R, SREBP1c transcription activity was significantly increased in mouse hepatocytes. CONCLUSION Taken together, our results demonstrate that disruption of A2A R in both macrophage and hepatocytes accounts for increased severity of NAFLD, likely through increasing inflammation and through elevating lipogenic events due to stimulation of SREBP1c expression and transcription activity. (Hepatology 2018;68:48-61).
Collapse
Affiliation(s)
- Yuli Cai
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA,Department of Endocrinology, Renmin Hospital, Wuhan University, Wuhan, Hubei 430060, China
| | - Honggui Li
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Mengyang Liu
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Ya Pei
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Juan Zheng
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA,Department of Endocrinology, Union Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jing Zhou
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Xianjun Luo
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Wenya Huang
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Linqiang Ma
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA,Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China,Laboratory of Lipid & Glucose Metabolism, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Qiuhua Yang
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA,Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Shaodong Guo
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Xiaoqiu Xiao
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China,Laboratory of Lipid & Glucose Metabolism, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Qifu Li
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Tianshu Zeng
- Department of Endocrinology, Union Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Fanyin Meng
- Research, Central Texas Veterans Health Care System, Texas A&M University College of Medicine, Temple, TX 76504, USA,Department of Medical Physiology, Texas A&M University College of Medicine, Temple, TX 76504, USA
| | - Heather Francis
- Research, Central Texas Veterans Health Care System, Texas A&M University College of Medicine, Temple, TX 76504, USA,Department of Medical Physiology, Texas A&M University College of Medicine, Temple, TX 76504, USA
| | - Shannon Glaser
- Research, Central Texas Veterans Health Care System, Texas A&M University College of Medicine, Temple, TX 76504, USA,Department of Medical Physiology, Texas A&M University College of Medicine, Temple, TX 76504, USA
| | - Lulu Chen
- Department of Endocrinology, Union Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Yuqing Huo
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA,Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Gianfranco Alpini
- Research, Central Texas Veterans Health Care System, Texas A&M University College of Medicine, Temple, TX 76504, USA,Department of Medical Physiology, Texas A&M University College of Medicine, Temple, TX 76504, USA,Contact information: Chaodong Wu, MD, PhD, College Station, TX 77843, Fax: 979 458 3129, ; or Gianfranco Alpini, PhD, Temple, TX 76504, ; Tel: 254 743 1041
| | - Chaodong Wu
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA,Contact information: Chaodong Wu, MD, PhD, College Station, TX 77843, Fax: 979 458 3129, ; or Gianfranco Alpini, PhD, Temple, TX 76504, ; Tel: 254 743 1041
| |
Collapse
|
23
|
Douros JD, Baltzegar DA, Reading BJ, Seale AP, Lerner DT, Grau EG, Borski RJ. Leptin Stimulates Cellular Glycolysis Through a STAT3 Dependent Mechanism in Tilapia. Front Endocrinol (Lausanne) 2018; 9:465. [PMID: 30186233 PMCID: PMC6110908 DOI: 10.3389/fendo.2018.00465] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 07/27/2018] [Indexed: 12/18/2022] Open
Abstract
We assessed if leptin, a cytokine hormone known to enhance energy expenditure by promoting lipid and carbohydrate catabolism in response to physiologic stress, might directly regulate cellular glycolysis. A transcriptomic analysis of prolactin cells in the tilapia (Oreochromis mossambicus) pituitary rostral pars distalis (RPD) revealed that recombinant leptin (rtLep) differentially regulates 1,995 genes, in vitro. Machine learning algorithms and clustering analyses show leptin influences numerous cellular gene networks including metabolism; protein processing, transport, and metabolism; cell cycle and the hypoxia response. Leptin stimulates transcript abundance of the glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase (gapdh) in a covariate manner to the hypoxic stress gene network. Orthogonal tests confirm that rtLepA dose-dependently increases gapdh gene expression in the RPD along with transcript abundance of 6-phosphofructo-1-kinase (pfk1), the rate limiting glycolytic enzyme. Functional testing demonstrated that leptin stimulates PFK activity and glycolytic output, while Stattic (a STAT3 blocker) was sufficient to suppress these responses, indicating leptin stimulates glycolysis through a STAT3-dependent mechanism. Leptin also stimulated pfk1 gene expression and lactate production in primary hepatocyte incubations in a similar manner to those shown for the pituitary RPD. This work characterizes a critical metabolic action of leptin to directly stimulate glycolysis across tissue types in a teleost model system, and suggest that leptin may promote energy expenditure, in part, by stimulating glycolysis. These data in a teleost fish, suggest that one of leptin's ancient, highly-conserved functions among vertebrates may be stimulation of glycolysis to facilitate the energetic needs associated with various stressors.
Collapse
Affiliation(s)
- Jonathan D. Douros
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| | - David A. Baltzegar
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
- Genomics Sciences Laboratory, North Carolina State University, Raleigh, NC, United States
| | - Benjamin J. Reading
- Department of Applied Ecology, North Carolina State University, Raleigh, NC, United States
| | - Andre P. Seale
- Hawaii Institute of Marine Biology, University of Hawaii, Kaneohe, HI, United States
- Department of Human Nutrition, Food, and Animal Sciences, University of Hawaii at Mānoa, Honolulu, HI, United States
| | - Darren T. Lerner
- Hawaii Institute of Marine Biology, University of Hawaii, Kaneohe, HI, United States
- University of Hawaii Sea Grant College Program, Honolulu, HI, United States
| | - E. Gordon Grau
- Hawaii Institute of Marine Biology, University of Hawaii, Kaneohe, HI, United States
| | - Russell J. Borski
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
- *Correspondence: Russell J. Borski
| |
Collapse
|
24
|
KO of 5-InsP 7 kinase activity transforms the HCT116 colon cancer cell line into a hypermetabolic, growth-inhibited phenotype. Proc Natl Acad Sci U S A 2017; 114:11968-11973. [PMID: 29078269 DOI: 10.1073/pnas.1702370114] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The inositol pyrophosphates 5-InsP7 (diphosphoinositol pentakisphosphate) and 1,5-InsP8 (bis-diphosphoinositol tetrakisphosphate) are highly energetic cellular signals interconverted by the diphosphoinositol pentakisphosphate kinases (PPIP5Ks). Here, we used CRISPR to KO PPIP5Ks in the HCT116 colon cancer cell line. This procedure eliminates 1,5-InsP8 and raises 5-InsP7 levels threefold. Expression of p53 and p21 was up-regulated; proliferation and G1/S cell-cycle transition slowed. Thus, PPIP5Ks are potential targets for tumor therapy. Deletion of the PPIP5Ks elevated [ATP] by 35%; both [ATP] and [5-InsP7] were restored to WT levels by overexpression of PPIP5K1, and a kinase-compromised PPIP5K1 mutant had no effect. This covariance of [ATP] with [5-InsP7] provides direct support for an energy-sensing attribute (i.e., 1 mM Km for ATP) of the 5-InsP7-generating inositol hexakisphosphate kinases (IP6Ks). We consolidate this conclusion by showing that 5-InsP7 levels are elevated on direct delivery of ATP into HCT116 cells using liposomes. Elevated [ATP] in PPIP5K-/- HCT116 cells is underpinned by increased mitochondrial oxidative phosphorylation and enhanced glycolysis. To distinguish between 1,5-InsP8 and 5-InsP7 as drivers of the hypermetabolic and p53-elevated phenotypes, we used IP6K2 RNAi and the pan-IP6K inhibitor, N2-(m-trifluorobenzyl), N6-(p-nitrobenzyl) purine (TNP), to return 5-InsP7 levels in PPIP5K-/- cells to those of WT cells without rescuing 1,5-InsP8 levels. Attenuation of IP6K restored p53 expression but did not affect the hypermetabolic phenotype. Thus, we conclude that 5-InsP7 regulates p53 expression, whereas 1,5-InsP8 regulates ATP levels. These findings attribute hitherto unsuspected functionality for 1,5-InsP8 to bioenergetic homeostasis.
Collapse
|
25
|
Uchiwa T, Takai Y, Tashiro A, Furuse M, Yasuo S. Exposure of C57BL/6J mice to long photoperiod during early life stages increases body weight and alters plasma metabolomic profiles in adulthood. Physiol Rep 2017; 4:4/18/e12974. [PMID: 27650252 PMCID: PMC5037922 DOI: 10.14814/phy2.12974] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 08/24/2016] [Indexed: 01/21/2023] Open
Abstract
Perinatal photoperiod is an important regulator of physiological phenotype in adulthood. In this study, we demonstrated that postnatal (0–4 weeks old) exposure of C57BL/6J mice to long photoperiod induced persistent increase in body weight until adulthood, compared with the mice maintained under short photoperiod. The expression of peroxisome proliferator‐activated receptor δ, a gene involved in fatty acid metabolism, was decreased in 10‐week‐old mice exposed to long photoperiod during 0–4 or 4–8 weeks of age. Plasma metabolomic profiles of adult mice exposed to a long photoperiod during the postnatal period (0–4 LD) were compared to those in the mice exposed to short photoperiod during the same period. Cluster analysis revealed that both carbon metabolic pathway and nucleic acid pathway were altered by the postnatal photoperiod. Levels of metabolites involved in glycolysis were significantly upregulated in 0–4 LD, suggesting that the mice in 0–4 LD use the glycolytic pathway for energy expenditure rather than the fatty acid oxidation pathway. In addition, the mice in 0–4 LD exhibited high levels of purine metabolites, which have a role in neuroprotection. In conclusion, postnatal exposure of C57BL/6J mice to long photoperiod induces increase in body weight and various changes in plasma metabolic profiles during adulthood.
Collapse
Affiliation(s)
- Tatsuhiro Uchiwa
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | - Yusuke Takai
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | - Ayako Tashiro
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | - Mitsuhiro Furuse
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | - Shinobu Yasuo
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| |
Collapse
|
26
|
Hudoyo AW, Hirase T, Tandelillin A, Honda M, Shirai M, Cheng J, Morisaki H, Morisaki T. Role of AMPD2 in impaired glucose tolerance induced by high fructose diet. Mol Genet Metab Rep 2017; 13:23-29. [PMID: 28765812 PMCID: PMC5527155 DOI: 10.1016/j.ymgmr.2017.07.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/13/2017] [Accepted: 07/13/2017] [Indexed: 12/13/2022] Open
Abstract
A high intake of products containing fructose is known to mediate insulin resistance. In the liver, AMPD2, an isoform of AMPD, has important glucose metabolic homeostasis functions including maintenance of AMP-activated protein kinase (AMPK). We speculated that AMPD2 induces impaired glucose tolerance in individuals who consume a high-fructose diet. We gave either a normal-chow (NCD) or high-fructose (HFrD) diet for 40 days to 8-week-old male wild-type (WT) and Ampd2 −/− homozygote (A2 −/−) C57BL/6 mice. A glucose tolerance test (GTT) and pyruvate tolerance test (PTT) were used to evaluate glucose metabolism. In addition, gluconeogenesis and glycolysis enzymes, and AMPK phosphorylation in the liver were investigated. With consumption of the HFrD, A2 −/− mice showed enhanced glucose tolerance in GTT and PTT results as compared to the WT mice, which were independent of changes in body weight. Also, the levels of phosphoenolpyruvate carboxy kinase and glucose-6-phosphatase (hepatic gluconeogenic enzymes) were significantly reduced in A2 −/− as compared to WT mice. The hepatic glycolytic enzymes glucokinase, phosphofructokinase, and pyruvate kinase were also examined, though there were no significant differences between genotypes in regard to both mRNA expression and protein expression under HFrD. Surprisingly, hepatic AMPK phosphorylation resulted in no changes in the A2 −/− as compared to WT mice under these conditions. Our results indicated that Ampd2–deficient mice are protected from high fructose diet-induced glycemic dysregulation, mainly because of gluconeogenesis inhibition, and indicate a novel therapeutic target for type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Athanasius Wrin Hudoyo
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Tetsuaki Hirase
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Andreas Tandelillin
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Masahiko Honda
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Manabu Shirai
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Jidong Cheng
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Hiroko Morisaki
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Takayuki Morisaki
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan.,Tokyo University of Technology School of Health Sciences, Ota, Tokyo, Japan
| |
Collapse
|
27
|
Kimura T, Kanasaki A, Hayashi N, Yamada T, Iida T, Nagata Y, Okuma K. d-Allulose enhances postprandial fat oxidation in healthy humans. Nutrition 2017; 43-44:16-20. [PMID: 28935140 DOI: 10.1016/j.nut.2017.06.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/28/2017] [Accepted: 06/29/2017] [Indexed: 11/28/2022]
Abstract
OBJECTIVE d-Allulose, a C-3 epimer of d-fructose, has been reported to decrease body weight and adipose tissue weight in animal studies and is expected to be a potent antiobese sweetener. Our animal study suggested that one of the mechanisms of d-allulose's antiobesity function is an increase in energy expenditure. However, a few studies have thus far explored the underlying mechanism in humans. The aim of this study was to examine the effects of a single ingestion of d-allulose on postprandial energy metabolism in healthy participants. METHODS Thirteen healthy men and women (mean age of 35.7 ± 2.1 y and body mass index 20.9 ± 0.7 kg/m2) were studied. The study was a randomized, single-blind crossover design with a 1-wk washout period. At 30 min after taking 5 g of d-allulose or 10 mg of aspartame without any sugar as a control, overnight-fasted participants ingested a standardized meal, and energy metabolism was evaluated by a breath-by-breath method. During the experiment, blood was collected and biochemical parameters such as plasma glucose were analyzed. RESULTS In the d-allulose-treated group, the area under the curve of fat oxidation was significantly higher than in the control group (10.5 ± 0.4 versus 9.6 ± 0.3 kJ·4 h·kg-1 body weight [BW]; P < 0.05), whereas that of carbohydrate oxidation was significantly lower (8.1 ± 0.5 versus 9.2 ± 0.5 kJ·4 h·kg-1 BW; P < 0.05). Furthermore, plasma glucose levels were significantly lower, and free fatty acid levels were significantly higher in the d-allulose group than in the control group. No other parameters such as insulin, total cholesterol, or triacylglycerol were modified. CONCLUSION d-Allulose enhances postprandial fat oxidation in healthy humans, indicating that it could be a novel sweetener to control and maintain healthy body weight, probably through enhanced energy metabolism.
Collapse
Affiliation(s)
- Tomonori Kimura
- Research and Development Department, Matsutani Chemical Industry Co., Ltd., Hyogo, Japan.
| | - Akane Kanasaki
- Research and Development Department, Matsutani Chemical Industry Co., Ltd., Hyogo, Japan
| | - Noriko Hayashi
- Research and Development Department, Matsutani Chemical Industry Co., Ltd., Hyogo, Japan
| | - Takako Yamada
- Research and Development Department, Matsutani Chemical Industry Co., Ltd., Hyogo, Japan
| | - Tetsuo Iida
- Research and Development Department, Matsutani Chemical Industry Co., Ltd., Hyogo, Japan
| | - Yasuo Nagata
- Center for Industry, University and Government Cooperation, Nagasaki University, Nagasaki, Japan
| | - Kazuhiro Okuma
- Research and Development Department, Matsutani Chemical Industry Co., Ltd., Hyogo, Japan
| |
Collapse
|
28
|
Qi T, Chen Y, Li H, Pei Y, Woo SL, Guo X, Zhao J, Qian X, Awika J, Huo Y, Wu C. A role for PFKFB3/iPFK2 in metformin suppression of adipocyte inflammatory responses. J Mol Endocrinol 2017; 59:49-59. [PMID: 28559290 PMCID: PMC5512603 DOI: 10.1530/jme-17-0066] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 04/20/2017] [Indexed: 12/16/2022]
Abstract
Metformin improves obesity-associated metabolic dysregulation, but has controversial effects on adipose tissue inflammation. The objective of the study is to examine the direct effect of metformin on adipocyte inflammatory responses and elucidate the underlying mechanisms. Adipocytes were differentiated from 3T3-L1 cells and treated with metformin at various doses and for different time periods. The treated cells were examined for the proinflammatory responses, as well as the phosphorylation states of AMPK and the expression of PFKFB3/iPFK2. In addition, PFKFB3/iPFK2-knockdown adipocytes were treated with metformin and examined for changes in the proinflammatory responses. The following results were obtained from the study. Treatment of adipocytes with metformin decreased the effects of lipopolysaccharide on inducing the phosphorylation states of JNK p46 and on increasing the mRNA levels of IL-1β and TNFα. In addition, treatment with metformin increased the expression of PFKFB3/iPFK2, but failed to significantly alter the phosphorylation states of AMPK. In PFKFB3/iPFK2-knockdown adipocytes, treatment with metformin did not suppress the proinflammatory responses as did it in control adipocytes. In conclusion, metformin has a direct effect on suppressing adipocyte proinflammatory responses in an AMPK-independent manner. Also, metformin increases adipocyte expression of PFKFB3/iPFK2, which is involved in the anti-inflammatory effect of metformin.
Collapse
Affiliation(s)
- Ting Qi
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
| | - Yanming Chen
- Department of Endocrinologythe Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Honggui Li
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
| | - Ya Pei
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
| | - Shih-Lung Woo
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
| | - Xin Guo
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
| | - Jiajia Zhao
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
| | - Xiaoxian Qian
- Department of Cardiologythe Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Joseph Awika
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
| | - Yuqing Huo
- Vascular Biology CenterDepartment of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, USA
- Drug Discovery CenterKey Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Chaodong Wu
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
| |
Collapse
|
29
|
Quines CB, Rosa SG, Chagas PM, Velasquez D, Prado VC, Nogueira CW. (p-ClPhSe) 2 stimulates carbohydrate metabolism and reverses the metabolic alterations induced by high fructose load in rats. Food Chem Toxicol 2017; 107:122-128. [PMID: 28655652 DOI: 10.1016/j.fct.2017.06.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 06/21/2017] [Accepted: 06/22/2017] [Indexed: 02/07/2023]
Abstract
The modern life leads to excess consumption of food rich in fructose; however, the long-term changes in carbohydrate and lipid metabolism could lead to metabolic dysfunction in humans. The present study evaluated the in vitro insulin-mimetic action of p-chloro-diphenyl diselenide (p-ClPhSe)2. The second aim of this study was to investigate if (p-ClPhSe)2 reverses metabolic dysfunction induced by fructose load in Wistar rats. The insulin-mimetic action of (p-ClPhSe)2 at concentrations of 50 and 100 μM was determined in slices of rat skeletal muscle. (p-ClPhSe)2 at a concentration of 50 μM stimulated the glucose uptake by 40% in skeletal muscle. A dose-response curve revealed that (p-ClPhSe)2 at a dose of 25 mg/kg reduced (∼20%) glycemia in rats treated with fructose (5 g/kg, i.g.). The administration of fructose impaired the liver homeostasis and (p-ClPhSe)2 (25 mg/kg) protected against the increase (∼25%) in the G-6-Pase and isocitrate dehydrogenase activities and reduced the triglyceride content (∼25%) in the liver. (p-ClPhSe)2 regulated the liver homeostasis by stimulating hexokinase activity (∼27%), regulating the TCA cycle activity (increased the ATP and citrate synthase activity (∼15%)) and increasing the glycogen levels (∼67%). In conclusion, (p-ClPhSe)2 stimulated carbohydrate metabolism and reversed metabolic dysfunction in rats fed with fructose.
Collapse
Affiliation(s)
- Caroline B Quines
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria, Santa Maria CEP 97105-900, RS, Brazil
| | - Suzan G Rosa
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria, Santa Maria CEP 97105-900, RS, Brazil
| | - Pietro M Chagas
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria, Santa Maria CEP 97105-900, RS, Brazil
| | - Daniela Velasquez
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria, Santa Maria CEP 97105-900, RS, Brazil
| | - Vinicius C Prado
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria, Santa Maria CEP 97105-900, RS, Brazil
| | - Cristina W Nogueira
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria, Santa Maria CEP 97105-900, RS, Brazil.
| |
Collapse
|
30
|
Richards P, Ourabah S, Montagne J, Burnol AF, Postic C, Guilmeau S. MondoA/ChREBP: The usual suspects of transcriptional glucose sensing; Implication in pathophysiology. Metabolism 2017; 70:133-151. [PMID: 28403938 DOI: 10.1016/j.metabol.2017.01.033] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 01/21/2017] [Indexed: 12/22/2022]
Abstract
Identification of the Mondo glucose-responsive transcription factors family, including the MondoA and MondoB/ChREBP paralogs, has shed light on the mechanism whereby glucose affects gene transcription. They have clearly emerged, in recent years, as key mediators of glucose sensing by multiple cell types. MondoA and ChREBP have overlapping yet distinct expression profiles, which underlie their downstream targets and separate roles in regulating genes involved in glucose metabolism. MondoA can restrict glucose uptake and influences energy utilization in skeletal muscle, while ChREBP signals energy storage through de novo lipogenesis in liver and white adipose tissue. Because Mondo proteins mediate metabolic adaptations to changing glucose levels, a better understanding of cellular glucose sensing through Mondo proteins will likely uncover new therapeutic opportunities in the context of the imbalanced glucose homeostasis that accompanies metabolic diseases such as type 2 diabetes and cancer. Here, we provide an overview of structural homologies, transcriptional partners as well as the nutrient and hormonal mechanisms underlying Mondo proteins regulation. We next summarize their relative contribution to energy metabolism changes in physiological states and the evolutionary conservation of these pathways. Finally, we discuss their possible targeting in human pathologies.
Collapse
Affiliation(s)
- Paul Richards
- Inserm, U1016, Institut Cochin, Paris, 75014, France; CNRS, UMR 8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Sarah Ourabah
- Inserm, U1016, Institut Cochin, Paris, 75014, France; CNRS, UMR 8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jacques Montagne
- Institut for Integrative Biology of the Cell (I2BC), CNRS, Université Paris-Sud, CEA, UMR 9198, F-91190, Gif-sur-Yvette, France
| | - Anne-Françoise Burnol
- Inserm, U1016, Institut Cochin, Paris, 75014, France; CNRS, UMR 8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Catherine Postic
- Inserm, U1016, Institut Cochin, Paris, 75014, France; CNRS, UMR 8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Sandra Guilmeau
- Inserm, U1016, Institut Cochin, Paris, 75014, France; CNRS, UMR 8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
31
|
Shintani T, Yamada T, Hayashi N, Iida T, Nagata Y, Ozaki N, Toyoda Y. Rare Sugar Syrup Containing d-Allulose but Not High-Fructose Corn Syrup Maintains Glucose Tolerance and Insulin Sensitivity Partly via Hepatic Glucokinase Translocation in Wistar Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:2888-2894. [PMID: 28209058 DOI: 10.1021/acs.jafc.6b05627] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Ingestion of high-fructose corn syrup (HFCS) is associated with the risk of both diabetes and obesity. Rare sugar syrup (RSS) has been developed by alkaline isomerization of HFCS and has anti-obesity and anti-diabetic effects. However, the influence of RSS on glucose metabolism has not been explored. We investigated whether long-term administration of RSS maintains glucose tolerance and whether the underlying mechanism involves hepatic glucokinase translocation. Wistar rats were administered water, RSS, or HFCS in drinking water for 10 weeks and then evaluated for glucose tolerance, insulin tolerance, liver glycogen content, and subcellular distribution of liver glucokinase. RSS significantly suppressed body weight gain and abdominal fat mass (p < 0.05). The glucose tolerance test revealed significantly higher blood glucose levels in the HFCS group compared to the water group, whereas the RSS group had significantly lower blood glucose levels from 90 to 180 min (p < 0.05). At 30, 60, and 90 min, the levels of insulin in the RSS group were significantly lower than those in the water group (p < 0.05). The amount of hepatic glycogen was more than 3 times higher in the RSS group than that in the other groups. After glucose loading, the nuclear export of glucokinase was significantly increased in the RSS group compared to the water group. These results imply that RSS maintains glucose tolerance and insulin sensitivity, at least partly, by enhancing nuclear export of hepatic glucokinase.
Collapse
Affiliation(s)
- Tomoya Shintani
- Research and Development, Matsutani Chemical Industry Company, Limited , 5-3 Kita-Itami, Itami, Hyogo 664-8508, Japan
| | - Takako Yamada
- Research and Development, Matsutani Chemical Industry Company, Limited , 5-3 Kita-Itami, Itami, Hyogo 664-8508, Japan
| | - Noriko Hayashi
- Research and Development, Matsutani Chemical Industry Company, Limited , 5-3 Kita-Itami, Itami, Hyogo 664-8508, Japan
| | - Tetsuo Iida
- Research and Development, Matsutani Chemical Industry Company, Limited , 5-3 Kita-Itami, Itami, Hyogo 664-8508, Japan
| | - Yasuo Nagata
- Center for Industry, University and Government Cooperation, Nagasaki University , 1-14 Bunkyo-machi, Nagasaki 852-9521, Japan
| | - Nobuaki Ozaki
- Japanese Red Cross Nagoya Daiichi Hospital , 3-35 Michishita, Nakamuraku-ku, Nagoya, Aichi 453-8511, Japan
| | - Yukiyasu Toyoda
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University , 150 Yagotoyama, Tempaku-ku, Nagoya, Aichi468-8503, Japan
| |
Collapse
|
32
|
Berberine Ameliorates Hepatic Steatosis and Suppresses Liver and Adipose Tissue Inflammation in Mice with Diet-induced Obesity. Sci Rep 2016; 6:22612. [PMID: 26936230 PMCID: PMC4776174 DOI: 10.1038/srep22612] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 02/17/2016] [Indexed: 12/12/2022] Open
Abstract
Increasing evidence demonstrates that berberine (BBR) is beneficial for obesity-associated non-alcoholic fatty liver disease (NAFLD). However, it remains to be elucidated how BBR improves aspects of NAFLD. Here we revealed an AMP-activated protein kinase (AMPK)-independent mechanism for BBR to suppress obesity-associated inflammation and improve hepatic steatosis. In C57BL/6J mice fed a high-fat diet (HFD), treatment with BBR decreased inflammation in both the liver and adipose tissue as indicated by reduction of the phosphorylation state of JNK1 and the mRNA levels of proinflammatory cytokines. BBR treatment also decreased hepatic steatosis, as well as the expression of acetyl-CoA carboxylase and fatty acid synthase. Interestingly, treatment with BBR did not significantly alter the phosphorylation state of AMPK in both the liver and adipose tissue of HFD-fed mice. Consistently, BBR treatment significantly decreased the phosphorylation state of JNK1 in both hepatoma H4IIE cells and mouse primary hepatocytes in both dose-dependent and time-dependent manners, which was independent of AMPK phosphorylation. BBR treatment also caused a decrease in palmitate-induced fat deposition in primary mouse hepatocytes. Taken together, these results suggest that BBR actions on improving aspects of NAFLD are largely attributable to BBR suppression of inflammation, which is independent of AMPK.
Collapse
|
33
|
Blum Y, Houée-Bigot M, Causeur D. Sparse factor model for co-expression networks with an application using prior biological knowledge. Stat Appl Genet Mol Biol 2016; 15:253-72. [DOI: 10.1515/sagmb-2015-0002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstractInference on gene regulatory networks from high-throughput expression data turns out to be one of the main current challenges in systems biology. Such networks can be very insightful for the deep understanding of interactions between genes. Because genes-gene interactions is often viewed as joint contributions to known biological mechanisms, inference on the dependence among gene expressions is expected to be consistent to some extent with the functional characterization of genes which can be derived from ontologies (GO, KEGG, …). The present paper introduces a sparse factor model as a general framework either to account for a prior knowledge on joint contributions of modules of genes to latent biological processes or to infer on the corresponding co-expression network. We propose an
Collapse
|
34
|
Chang RCA, Shi L, Huang CCY, Kim AJ, Ko ML, Zhou B, Ko GYP. High-Fat Diet-Induced Retinal Dysfunction. Invest Ophthalmol Vis Sci 2015; 56:2367-80. [PMID: 25788653 DOI: 10.1167/iovs.14-16143] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
PURPOSE The purpose of this study was to investigate the impact of obesity-induced prediabetes/early diabetes on the retina to provide new evidence on the pathogenesis of type 2 diabetes-associated diabetic retinopathy (DR). METHODS A high-fat diet (HFD)-induced obesity mouse model (male C57BL/6J) was used in this study. At the end of the 12-week HFD feeding regimen, mice were evaluated for glucose and insulin tolerance, and retinal light responses were recorded by electroretinogram (ERG). Western immunoblot and immunohistochemical staining were used to determine changes in elements regulating calcium homeostasis between HFD and control retinas, as well as unstained human retinal sections from DR patients and age-appropriate controls. RESULTS Compared to the control, the scotopic and photopic ERGs from HFD mice were decreased. There were significant decreases in molecules related to cell signaling, calcium homeostasis, and glucose metabolism from HFD retinas, including phosphorylated protein kinase B (pAKT), glucose transporter 4, L-type voltage-gated calcium channel (L-VGCC), and plasma membrane calcium ATPase (PMCA). Similar changes for pAKT, PMCA, and L-VGCC were also observed in human retinal sections from DR patients. CONCLUSIONS Obesity-induced hyperglycemic and prediabetic/early diabetic conditions caused detrimental impacts on retinal light sensitivities and health. The decrease of the ERG components in early diabetes reflects the decreased neuronal activity of retinal light responses, which may be caused by a decrease in neuronal calcium signaling. Since PI3K-AKT is important in regulating calcium homeostasis and neural survival, maintaining proper PI3K-AKT signaling in early diabetes or at the prediabetic stage might be a new strategy for DR prevention.
Collapse
Affiliation(s)
- Richard Cheng-An Chang
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States
| | - Liheng Shi
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States
| | - Cathy Chia-Yu Huang
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States
| | - Andy Jeesu Kim
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States
| | - Michael L Ko
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States
| | - Beiyan Zhou
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States
| | - Gladys Y-P Ko
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States 3Texas A&M Institute of Neuroscience, Texas A&M University, College Station, Texas, Unite
| |
Collapse
|
35
|
Parray HA, Yun JW. Proteomic Identification of Target Proteins of Thiodigalactoside in White Adipose Tissue from Diet-Induced Obese Rats. Int J Mol Sci 2015; 16:14441-63. [PMID: 26121299 PMCID: PMC4519851 DOI: 10.3390/ijms160714441] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Revised: 06/15/2015] [Accepted: 06/18/2015] [Indexed: 12/11/2022] Open
Abstract
Previously, galectin-1 (GAL1) was found to be up-regulated in obesity-prone subjects, suggesting that use of a GAL1 inhibitor could be a novel therapeutic approach for treatment of obesity. We evaluated thiodigalactoside (TDG) as a potent inhibitor of GAL1 and identified target proteins of TDG by performing comparative proteome analysis of white adipose tissue (WAT) from control and TDG-treated rats fed a high fat diet (HFD) using two dimensional gel electrophoresis (2-DE) combined with MALDI-TOF-MS. Thirty-two spots from a total of 356 matched spots showed differential expression between control and TDG-treated rats, as identified by peptide mass fingerprinting. These proteins were categorized into groups such as carbohydrate metabolism, tricarboxylic acid (TCA) cycle, signal transduction, cytoskeletal, and mitochondrial proteins based on functional analysis using Protein Annotation Through Evolutionary Relationship (PANTHER) and Database for Annotation, Visualization, Integrated Discovery (DAVID) classification. One of the most striking findings of this study was significant changes in Carbonic anhydrase 3 (CA3), Voltage-dependent anion channel 1 (VDAC1), phosphatidylethanolamine-binding protein 1 (PEBP1), annexin A2 (ANXA2) and lactate dehydrogenase A chain (LDHA) protein levels between WAT from control and TDG-treated groups. In addition, we confirmed increased expression of thermogenic proteins as well as reduced expression of lipogenic proteins in response to TDG treatment. These results suggest that TDG may effectively prevent obesity, and TDG-responsive proteins can be used as novel target proteins for obesity treatment.
Collapse
Affiliation(s)
- Hilal Ahmad Parray
- Department of Biotechnology, Daegu University, Kyungsan, Kyungbuk 712-714, Korea.
| | - Jong Won Yun
- Department of Biotechnology, Daegu University, Kyungsan, Kyungbuk 712-714, Korea.
| |
Collapse
|
36
|
Luo Y, Dong X, Yu Y, Sun G, Sun X. Total aralosides of aralia elata (Miq) seem (TASAES) ameliorate nonalcoholic steatohepatitis by modulating IRE1α-mediated JNK and NF-κB pathways in ApoE-/- mice. JOURNAL OF ETHNOPHARMACOLOGY 2015; 163:241-250. [PMID: 25655997 DOI: 10.1016/j.jep.2015.01.017] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 12/10/2014] [Accepted: 01/15/2015] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Total saponins of Aralia elata (Miq) Seem (TASAES) from the Chinese traditional herb Long ya Aralia chinensis L. is popularly used as a folk medicine to treat rheumatism, neurasthenia, diabetes, hepatitis and antivirus in Asian countries. However, there was poor study of TASAES on Non-alcoholic steatohepatitis (NASH), which is characterized by inflammatory responses and hepatocellular apoptosis exacerbating liver injury. This study aimed to clarify whether or not the anti-inflammatory and anti-apoptotic activities and protective mechanisms of the total aralosides of Aralia elata (Miq) Seem (TASAES) ameliorate NASH in a high-fat diet (HFD)-induced ApoE-/- mouse model. MATERIAL AND METHODS C57/BL6N and ApoE-/- mice were fed with HFD containing 0.3% cholesterol and 20% fat to induce NASH and then treated with TASAES (75,150mg/kg/day, i.g.) for 12 weeks. Liver tissue was procured for histological examination, real-time RT-PCR and Western blot analysis. RESULTS ASAES treatment groups exhibited lower serum alanine and aspartate aminotransferases than the NASH group. TASAES could also reduce hepatic steatosis, as revealed by histological changes. In addition, TASAES treatment groups showed lower protein and mRNA expression levels of pro-inflammatory cytokines, such as IL-6, MCP-1, and TNF-α than NASH group. Reduced TUNEL-positive cells were also found in TASAES treatment groups. Western blot and immunohistochemical results indicated that TASAES regulated apoptosis and inflammation-related protein expression. Furthermore, TASAES treatment significantly reduced the phosphorylation of IRE1α, JNK and IκB and the downstream activation of NF-κB p65 was also reduced. CONCLUSION These findings suggested that the ameliorative effects of TASASE in HFD-induced NASH were associated with the regulation of IRE1α-mediated JNK and NF-κB signal pathways, thereby protecting the liver against NASH.
Collapse
Affiliation(s)
- Yun Luo
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, PR China
| | - Xi Dong
- Academy of Chinese Materia Medica, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yingli Yu
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, PR China
| | - Guibo Sun
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, PR China.
| | - Xiaobo Sun
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, PR China.
| |
Collapse
|
37
|
Muller YL, Piaggi P, Hanson RL, Kobes S, Bhutta S, Abdussamad M, Leak-Johnson T, Kretzler M, Huang K, Weil EJ, Nelson RG, Knowler WC, Bogardus C, Baier LJ. A cis-eQTL in PFKFB2 is associated with diabetic nephropathy, adiposity and insulin secretion in American Indians. Hum Mol Genet 2015; 24:2985-96. [PMID: 25662186 DOI: 10.1093/hmg/ddv040] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 02/02/2015] [Indexed: 01/13/2023] Open
Abstract
A prior genome-wide association study (GWAS) in Pima Indians identified a variant within PFKFB2 (rs17258746) associated with body mass index (BMI). PFKFB2 encodes 6-phosphofructo-2-kinase/fructose 2,6-bisphosphatase isoform 2, which plays a role in glucose metabolism. To follow-up on the GWAS, tag SNPs across PFKFB2 were genotyped in American Indians (AI) who had longitudinal data on BMI (n = 6839), type 2 diabetes (T2D; n = 7710), diabetic nephropathy (DN; n = 2452), % body fat (n = 555) and insulin secretion (n = 298). Two SNPs were further genotyped in urban AI to assess replication for DN (n = 864). PFKFB2 expression was measured in 201 adipose biopsies using real-time RT-PCR and 61 kidney biopsies using the Affymetrix U133 array. Two SNPs (rs17258746 and rs11120137), which capture the same signal, were associated with maximum BMI in adulthood (β = 1.02 per risk allele, P = 7.3 × 10(-4)), maximum BMI z-score in childhood (β = 0.079, P = 0.03) and % body fat in adulthood (β = 3.4%, P = 3 × 10(-7)). The adiposity-increasing allele correlated with lower PFKFB2 adipose expression (β = 0.81, P = 9.4 × 10(-4)). Lower expression of PFKFB2 further correlated with higher % body fat (r = -0.16, P = 0.02) and BMI (r = -0.17, P = 0.02). This allele was also associated with increased risk for DN in both cohorts of AI [odds ratio = 1.64 (1.32-2.02), P = 5.8 × 10(-6)], and similarly correlated with lower PFKFB2 expression in kidney glomeruli (β = 0.87, P = 0.03). The same allele was also associated with lower insulin secretion assessed by acute insulin response (β = 0.78, P = 0.03) and 30-min plasma insulin concentrations (β = 0.78, P = 1.1 × 10(-4)). Variation in PFKFB2 appears to reduce PFKFB2 expression in adipose and kidney tissues, and thereby increase risk for adiposity and DN.
Collapse
Affiliation(s)
- Yunhua L Muller
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, Phoenix, AZ, USA and
| | - Paolo Piaggi
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, Phoenix, AZ, USA and
| | - Robert L Hanson
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, Phoenix, AZ, USA and
| | - Sayuko Kobes
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, Phoenix, AZ, USA and
| | - Shujera Bhutta
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, Phoenix, AZ, USA and
| | - Maryam Abdussamad
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, Phoenix, AZ, USA and
| | - Tennille Leak-Johnson
- Department of Internal Medicine and Computational Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Matthias Kretzler
- Department of Internal Medicine and Computational Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Ke Huang
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, Phoenix, AZ, USA and
| | - E Jennifer Weil
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, Phoenix, AZ, USA and
| | - Robert G Nelson
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, Phoenix, AZ, USA and
| | - William C Knowler
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, Phoenix, AZ, USA and
| | - Clifton Bogardus
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, Phoenix, AZ, USA and
| | - Leslie J Baier
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, Phoenix, AZ, USA and
| |
Collapse
|
38
|
PFKFB4 controls embryonic patterning via Akt signalling independently of glycolysis. Nat Commun 2015; 6:5953. [PMID: 25601028 DOI: 10.1038/ncomms6953] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 11/26/2014] [Indexed: 12/25/2022] Open
Abstract
How metabolism regulators play roles during early development remains elusive. Here we show that PFKFB4 (6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 4), a glycolysis regulator, is critical for controlling dorsal ectoderm global patterning in gastrulating frog embryos via a non-glycolytic function. PFKFB4 is required for dorsal ectoderm progenitors to proceed towards more specified fates including neural and non-neural ectoderm, neural crest or placodes. This function is mediated by Akt signalling, a major pathway that integrates cell homeostasis and survival parameters. Restoring Akt signalling rescues the loss of PFKFB4 in vivo. In contrast, glycolysis is not essential for frog development at this stage. Our study reveals the existence of a PFKFB4-Akt checkpoint that links cell homeostasis to the ability of progenitor cells to undergo differentiation, and uncovers glycolysis-independent functions of PFKFB4.
Collapse
|
39
|
Liu S, Alexander RK, Lee CH. Lipid metabolites as metabolic messengers in inter-organ communication. Trends Endocrinol Metab 2014; 25:356-63. [PMID: 24895003 PMCID: PMC4077945 DOI: 10.1016/j.tem.2014.05.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Revised: 04/30/2014] [Accepted: 05/07/2014] [Indexed: 01/08/2023]
Abstract
Metabolic homeostasis is achieved through coordinated regulation across several tissues. Studies using mouse genetic models have shown that perturbation of specific pathways of lipid metabolism in metabolically active tissues impacts systemic metabolic homeostasis. The use of metabolomic technologies combined with genetic models has helped to identify several potential lipid mediators that serve as metabolic messengers to communicate energy status and modulate substrate utilization among tissues. When provided exogenously, these lipid metabolites exhibit biological effects on glucose and lipid metabolism, indicating a therapeutic potential for treating metabolic diseases. In this review we summarize recent advances in inter-organ communication through novel mechanisms, with a focus on lipid mediators synthesized de novo or derived from dietary sources, and discuss challenges and future directions.
Collapse
Affiliation(s)
- Sihao Liu
- Department of Genetics and Complex Diseases, Division of Biological Sciences, Harvard School of Public Health, Boston, MA 02115, USA
| | - Ryan K Alexander
- Department of Genetics and Complex Diseases, Division of Biological Sciences, Harvard School of Public Health, Boston, MA 02115, USA
| | - Chih-Hao Lee
- Department of Genetics and Complex Diseases, Division of Biological Sciences, Harvard School of Public Health, Boston, MA 02115, USA.
| |
Collapse
|
40
|
Shannonhouse JL, Urbanski HF, Woo SL, Fong LA, Goddard SD, Lucas WF, Jones ER, Wu C, Morgan C. Aquaporin-11 control of testicular fertility markers in Syrian hamsters. Mol Cell Endocrinol 2014; 391:1-9. [PMID: 24791736 PMCID: PMC4368057 DOI: 10.1016/j.mce.2014.04.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 02/14/2014] [Accepted: 04/17/2014] [Indexed: 10/25/2022]
Abstract
The present study sought novel changes to the hamster testicular transcriptome during modulation of fertility by well-characterized photoperiodic stimuli. Transition from long days (LD, 14 h light/day) to short days (SD, 10h light/day) triggered testicular regression (61% reduction of testis weight, relative to LD) in SD-sensitive (SD-S) hamsters within 16 weeks. After 22 weeks of SD exposure, a third cohort of hamsters became SD-refractory (SD-R), and exhibited testicular recrudescence (137% testis weight gain, relative to SD-S). Partial interrogation of the testicular transcriptome by annealing-control-primer-modified differential display PCR provided several candidates for regulation of testicular functions. Multiple linear regression modeling indicated the best correlation for aquaporin 11 (Aqp11) with changes in testis weight. Correlations were also strongest for Aqp11 with expression levels of reference cDNAs that control spermatogenesis (Hspa2 and Tnp2), steroidogenesis (Cox2, 3βHsd, and Srebp2), sperm motility (Catsper1, Pgk2, and Tnp2), inflammation (Cox2), and apoptosis (Bax and Bcl2). Moreover, siRNA-mediated knockdown of testicular Aqp11 mRNA and protein reduced Hspa2 and Tnp2 mRNA levels, and it increased 3βHsd mRNA levels. It also reduced mRNA levels for Sept12, which is a testis-specific inducer of spermatogenesis. These results suggest a central role for testicular Aqp11 signaling in the coordinate regulation of crucial components of fertility.
Collapse
Affiliation(s)
- John L Shannonhouse
- Institute for Neuroscience, Texas A&M University, College Station, TX 77843, USA
| | - Henryk F Urbanski
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Shih-Lung Woo
- Department of Nutrition & Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Li An Fong
- Department of Nutrition & Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Scott D Goddard
- Department of Statistics, Texas A&M University, College Station, TX 77843, USA
| | - William F Lucas
- Department of Statistics, Texas A&M University, College Station, TX 77843, USA
| | - Edward R Jones
- Department of Statistics, Texas A&M University, College Station, TX 77843, USA
| | - Chaodong Wu
- Department of Nutrition & Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Caurnel Morgan
- Institute for Neuroscience, Texas A&M University, College Station, TX 77843, USA; Department of Nutrition & Food Science, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
41
|
Xu H, Li H, Woo SL, Kim SM, Shende VR, Neuendorff N, Guo X, Guo T, Qi T, Pei Y, Zhao Y, Hu X, Zhao J, Chen L, Chen L, Ji JY, Alaniz RC, Earnest DJ, Wu C. Myeloid cell-specific disruption of Period1 and Period2 exacerbates diet-induced inflammation and insulin resistance. J Biol Chem 2014; 289:16374-88. [PMID: 24770415 DOI: 10.1074/jbc.m113.539601] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The circadian clockworks gate macrophage inflammatory responses. Given the association between clock dysregulation and metabolic disorders, we conducted experiments to determine the extent to which over-nutrition modulates macrophage clock function and whether macrophage circadian dysregulation is a key factor linking over-nutrition to macrophage proinflammatory activation, adipose tissue inflammation, and systemic insulin resistance. Our results demonstrate that 1) macrophages from high fat diet-fed mice are marked by dysregulation of the molecular clockworks in conjunction with increased proinflammatory activation, 2) global disruption of the clock genes Period1 (Per1) and Per2 recapitulates this amplified macrophage proinflammatory activation, 3) adoptive transfer of Per1/2-disrupted bone marrow cells into wild-type mice potentiates high fat diet-induced adipose and liver tissue inflammation and systemic insulin resistance, and 4) Per1/2-disrupted macrophages similarly exacerbate inflammatory responses and decrease insulin sensitivity in co-cultured adipocytes in vitro. Furthermore, PPARγ levels are decreased in Per1/2-disrupted macrophages and PPARγ2 overexpression ameliorates Per1/2 disruption-associated macrophage proinflammatory activation, suggesting that this transcription factor may link the molecular clockworks to signaling pathways regulating macrophage polarization. Thus, macrophage circadian clock dysregulation is a key process in the physiological cascade by which diet-induced obesity triggers macrophage proinflammatory activation, adipose tissue inflammation, and insulin resistance.
Collapse
Affiliation(s)
- Hang Xu
- From the Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843
| | - Honggui Li
- From the Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843
| | - Shih-Lung Woo
- From the Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843
| | - Sam-Moon Kim
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, Texas 77807
| | - Vikram R Shende
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, Texas 77807
| | - Nichole Neuendorff
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, Texas 77807
| | - Xin Guo
- From the Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843
| | - Ting Guo
- From the Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843
| | - Ting Qi
- From the Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843
| | - Ya Pei
- From the Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843
| | - Yan Zhao
- From the Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843
| | - Xiang Hu
- From the Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843, Department of Endocrinology and
| | - Jiajia Zhao
- From the Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843, Department of Stomatology, Union Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China, and
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China, and
| | | | - Jun-Yuan Ji
- Department of Molecular and Cellular Medicine and
| | - Robert C Alaniz
- Department of Microbial and Molecular Pathogenesis, College of Medicine, Texas A&M Health Science Center, College Station, Texas 77843
| | - David J Earnest
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, Texas 77807,
| | - Chaodong Wu
- From the Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843,
| |
Collapse
|
42
|
Woo SL, Xu H, Li H, Zhao Y, Hu X, Zhao J, Guo X, Guo T, Botchlett R, Qi T, Pei Y, Zheng J, Xu Y, An X, Chen L, Chen L, Li Q, Xiao X, Huo Y, Wu C. Metformin ameliorates hepatic steatosis and inflammation without altering adipose phenotype in diet-induced obesity. PLoS One 2014; 9:e91111. [PMID: 24638078 PMCID: PMC3956460 DOI: 10.1371/journal.pone.0091111] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 02/06/2014] [Indexed: 01/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is closely associated with obesity and insulin resistance. To better understand the pathophysiology of obesity-associated NAFLD, the present study examined the involvement of liver and adipose tissues in metformin actions on reducing hepatic steatosis and inflammation during obesity. C57BL/6J mice were fed a high-fat diet (HFD) for 12 weeks to induce obesity-associated NAFLD and treated with metformin (150 mg/kg/d) orally for the last four weeks of HFD feeding. Compared with HFD-fed control mice, metformin-treated mice showed improvement in both glucose tolerance and insulin sensitivity. Also, metformin treatment caused a significant decrease in liver weight, but not adiposity. As indicated by histological changes, metformin treatment decreased hepatic steatosis, but not the size of adipocytes. In addition, metformin treatment caused an increase in the phosphorylation of liver AMP-activated protein kinase (AMPK), which was accompanied by an increase in the phosphorylation of liver acetyl-CoA carboxylase and decreases in the phosphorylation of liver c-Jun N-terminal kinase 1 (JNK1) and in the mRNA levels of lipogenic enzymes and proinflammatory cytokines. However, metformin treatment did not significantly alter adipose tissue AMPK phosphorylation and inflammatory responses. In cultured hepatocytes, metformin treatment increased AMPK phosphorylation and decreased fat deposition and inflammatory responses. Additionally, in bone marrow-derived macrophages, metformin treatment partially blunted the effects of lipopolysaccharide on inducing the phosphorylation of JNK1 and nuclear factor kappa B (NF-κB) p65 and on increasing the mRNA levels of proinflammatory cytokines. Taken together, these results suggest that metformin protects against obesity-associated NAFLD largely through direct effects on decreasing hepatocyte fat deposition and on inhibiting inflammatory responses in both hepatocytes and macrophages.
Collapse
Affiliation(s)
- Shih-Lung Woo
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America
| | - Hang Xu
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America
| | - Honggui Li
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America
| | - Yan Zhao
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America
| | - Xiang Hu
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America; Department of Endocrinology, Union Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiajia Zhao
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America; Department of Stomatology, Union Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xin Guo
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America
| | - Ting Guo
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America
| | - Rachel Botchlett
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America
| | - Ting Qi
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America
| | - Ya Pei
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America
| | - Juan Zheng
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America; Department of Endocrinology, Union Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yiming Xu
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States of America
| | - Xiaofei An
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Lulu Chen
- Department of Endocrinology, Union Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qifu Li
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoqiu Xiao
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Laboratory of Lipid & Glucose Metabolism, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuqing Huo
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States of America; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Chaodong Wu
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
43
|
Chronic exposure of mice to environmental endocrine-disrupting chemicals disturbs their energy metabolism. Toxicol Lett 2014; 225:392-400. [PMID: 24440342 DOI: 10.1016/j.toxlet.2014.01.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 01/04/2014] [Accepted: 01/07/2014] [Indexed: 11/23/2022]
Abstract
We evaluated the effects of a 20-week chronic exposure of mice to a low dose of cypermethrin (CYP), atrazine (ATZ) and 17α-ethynyestradiol (EE2) on energy metabolism. Here, male mice were exposed to 50 μg/kg BW/day CYP, 100 μg/kg BW/day ATZ or 1 μg/kg BW/day EE2 supplied in their drinking water for 20 weeks. During the exposure, mice were fed a high energy diet (HD). The bodyweights were not significantly affected by chronic exposure to EDCs, while the serum-free fatty acids (FFA) levels, hepatic lipid accumulation and triacylglycerol (TG) contents increased significantly in the ATZ- and CYP-HD groups. To determine the mechanism involved, we determined the expression levels of the genes in the glucose and fat metabolism pathways in the liver and adipose tissue. The results showed that chronic exposure to ATZ and CYP increased the mRNA levels of a number of key genes involved in both the de novo FFA synthesis pathway and the transport of FFA from blood. The increased amount of FFA was partially consumed as energy through β-oxidation in the mitochondria. Some of the FFA was used to synthesize TG in the liver by up-regulating primary genes, which resulted in increased TG levels and lipid accumulation. The results indicate that chronic exposure to EDCs has the potential to cause energy metabolic dysregulation and hepatotoxicity in mice.
Collapse
|
44
|
Yamada T, Tsukita S, Katagiri H. Identification of a novel interorgan mechanism favoring energy storage in overnutrition. Adipocyte 2013; 2:281-4. [PMID: 24052907 PMCID: PMC3774707 DOI: 10.4161/adip.25499] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 06/21/2013] [Accepted: 06/21/2013] [Indexed: 12/31/2022] Open
Abstract
While body weight is essentially determined by the balance of energy intake and energy consumption, it is not necessarily the case that changes in daily food intakes and exercise directly reflect changes in body weight. In recent years, it has been revealed that numerous metabolic interactions between organs, which are organized by the brain, function as a feedback mechanism, and are involved in maintaining body weight homeostasis against excess energy intake. On the other hand, since obesity has seen an explosive increase in this age of plenty, there must be other interactions between organs working as feedforward mechanisms favoring weight gain. However, no such interaction has yet been demonstrated. Recently, we discovered a new interorgan neural network, from the liver, which may represent the feedforward mechanism.(1) Under conditions of excessive energy intake, changes in glucose metabolism occur in the liver with increased expression of hepatic glucokinase (GK) and the induction of neuronal signal transmission via the afferent vagus nerve. These signals are received by the medulla and result in inactivation of sympathetic nerve to brown adipose tissue (BAT), thereby suppressing thermogenesis in BAT and promoting adiposity. Furthermore, the efficacy of the liver-to-BAT interaction differs among mouse strains and these differences may contribute to determining the obesity predispositions of various strains. In conclusion, this novel interorgan neuronal relay system functions to suppress energy expenditure when energy intake is increased, and thus, is considered to be a thrifty mechanism operating on the whole body level. During periods when sufficient food was not always available, this system worked in favor of survival. However, in the current age of plenty, it is assumed to work as a mechanism flipping a metabolic switch toward obesity.
Collapse
|
45
|
Filhoulaud G, Guilmeau S, Dentin R, Girard J, Postic C. Novel insights into ChREBP regulation and function. Trends Endocrinol Metab 2013; 24:257-68. [PMID: 23597489 DOI: 10.1016/j.tem.2013.01.003] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 12/23/2012] [Accepted: 01/03/2013] [Indexed: 12/17/2022]
Abstract
Glucose is an energy source that also controls the expression of key genes involved in energetic metabolism through the glucose-signaling transcription factor carbohydrate response element-binding protein (ChREBP). ChREBP has recently emerged as a central regulator of glycolysis and de novo fatty acid synthesis in liver, but new evidence shows that it plays a broader and crucial role in various processes, ranging from glucolipotoxicity to apoptosis and/or proliferation in specific cell types. However, several aspects of ChREBP activation by glucose metabolites are currently controversial, as well as the effects of activating or inhibiting ChREBP, on insulin sensitivity, which might depend on genetic, dietary or environmental factors. Thus, much remains to be elucidated. Here, we summarize our current understanding of the regulation and function of this fascinating transcription factor.
Collapse
|
46
|
Seyer P, Vallois D, Poitry-Yamate C, Schütz F, Metref S, Tarussio D, Maechler P, Staels B, Lanz B, Grueter R, Decaris J, Turner S, da Costa A, Preitner F, Minehira K, Foretz M, Thorens B. Hepatic glucose sensing is required to preserve β cell glucose competence. J Clin Invest 2013; 123:1662-76. [PMID: 23549084 DOI: 10.1172/jci65538] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 01/24/2013] [Indexed: 12/31/2022] Open
Abstract
Liver glucose metabolism plays a central role in glucose homeostasis and may also regulate feeding and energy expenditure. Here we assessed the impact of glucose transporter 2 (Glut2) gene inactivation in adult mouse liver (LG2KO mice). Loss of Glut2 suppressed hepatic glucose uptake but not glucose output. In the fasted state, expression of carbohydrate-responsive element-binding protein (ChREBP) and its glycolytic and lipogenic target genes was abnormally elevated. Feeding, energy expenditure, and insulin sensitivity were identical in LG2KO and control mice. Glucose tolerance was initially normal after Glut2 inactivation, but LG2KO mice exhibited progressive impairment of glucose-stimulated insulin secretion even though β cell mass and insulin content remained normal. Liver transcript profiling revealed a coordinated downregulation of cholesterol biosynthesis genes in LG2KO mice that was associated with reduced hepatic cholesterol in fasted mice and reduced bile acids (BAs) in feces, with a similar trend in plasma. We showed that chronic BAs or farnesoid X receptor (FXR) agonist treatment of primary islets increases glucose-stimulated insulin secretion, an effect not seen in islets from Fxr(-/-) mice. Collectively, our data show that glucose sensing by the liver controls β cell glucose competence and suggest BAs as a potential mechanistic link.
Collapse
Affiliation(s)
- Pascal Seyer
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Zhang F, Xu X, Zhang Y, Zhou B, He Z, Zhai Q. Gene expression profile analysis of type 2 diabetic mouse liver. PLoS One 2013; 8:e57766. [PMID: 23469233 PMCID: PMC3585940 DOI: 10.1371/journal.pone.0057766] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 01/24/2013] [Indexed: 01/18/2023] Open
Abstract
Liver plays a key role in glucose metabolism and homeostasis, and impaired hepatic glucose metabolism contributes to the development of type 2 diabetes. However, the precise gene expression profile of diabetic liver and its association with diabetes and related diseases are yet to be further elucidated. In this study, we detected the gene expression profile by high-throughput sequencing in 9-week-old normal and type 2 diabetic db/db mouse liver. Totally 12132 genes were detected, and 2627 genes were significantly changed in diabetic mouse liver. Biological process analysis showed that the upregulated genes in diabetic mouse liver were mainly enriched in metabolic processes. Surprisingly, the downregulated genes in diabetic mouse liver were mainly enriched in immune-related processes, although all the altered genes were still mainly enriched in metabolic processes. Similarly, KEGG pathway analysis showed that metabolic pathways were the major pathways altered in diabetic mouse liver, and downregulated genes were enriched in immune and cancer pathways. Analysis of the key enzyme genes in fatty acid and glucose metabolism showed that some key enzyme genes were significantly increased and none of the detected key enzyme genes were decreased. In addition, FunDo analysis showed that liver cancer and hepatitis were most likely to be associated with diabetes. Taken together, this study provides the digital gene expression profile of diabetic mouse liver, and demonstrates the main diabetes-associated hepatic biological processes, pathways, key enzyme genes in fatty acid and glucose metabolism and potential hepatic diseases.
Collapse
Affiliation(s)
- Fang Zhang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiang Xu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yi Zhang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ben Zhou
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhishui He
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qiwei Zhai
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- * E-mail:
| |
Collapse
|
48
|
Li H, Guo X, Xu H, Woo SL, Halim V, Morgan C, Wu C. A role for inducible 6-phosphofructo-2-kinase in the control of neuronal glycolysis. J Nutr Biochem 2012; 24:1153-8. [PMID: 23246158 DOI: 10.1016/j.jnutbio.2012.08.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 08/07/2012] [Accepted: 08/27/2012] [Indexed: 10/27/2022]
Abstract
Increased glycolysis is the result of the sensing of glucose by hypothalamic neurons. The biochemical mechanisms underlying the control of hypothalamic glycolysis, however, remain to be elucidated. Here we showed that PFKFB3, the gene that encodes for inducible 6-phosphofructo-2-kinase (iPFK2), was expressed at high abundance in both mouse hypothalami and clonal hypothalamic neurons. In response to re-feeding, PFKFB3 mRNA levels were increased by 10-fold in mouse hypothalami. In the hypothalamus, re-feeding also decreased the phosphorylation of AMP-activated protein kinase (AMPK) (Thr172) and the mRNA levels of agouti-related protein (AgRP), and increased the mRNA levels of cocaine-amphetamine-related transcript (CART). Similar results were observed in N-43/5 clonal hypothalamic neurons upon treatment with glucose and/or insulin. In addition, knockdown of PFKFB3/iPFK2 in N-43/5 neurons caused a decrease in rates of glycolysis, which was accompanied by increased AMPK phosphorylation, increased AgRP mRNA levels and decreased CART mRNA levels. In contrast, overexpression of PFKFB3/iPFK2 in N-43/5 neurons caused an increase in glycolysis, which was accompanied by decreased AMPK phosphorylation and decreased AgRP mRNA levels and increased CART mRNA levels. Together, these results suggest that PFKFB3/iPFK2 responds to re-feeding, which in turn stimulates hypothalamic glycolysis and decreases hypothalamic AMPK phosphorylation and alters neuropeptide expression in a pattern that is associated with suppression of food intake.
Collapse
Affiliation(s)
- Honggui Li
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Liver fructose-1,6-bisphosphatase (FBPase) is a regulatory enzyme in gluconeogenesis that is elevated by obesity and dietary fat intake. Whether FBPase functions only in glucose metabolism or has other metabolic roles is currently unclear. In our recently published study, we examined the importance of liver FBPase in body weight regulation by performing a series of comprehensive physiological and biochemical assessments of energy balance and specific intervention studies in our transgenic mouse line that overexpresses FBPase specifically in the liver. Compared with negative littermates, these FBPase transgenic mice weighed 10% less, had 50% less adiposity, ate 15% less food but did not have altered energy expenditure. Increased circulating leptin and cholecystokinin levels, elevated fatty acid oxidation and reduced appetite stimulating neuropeptides, neuropeptide Y (NPY) and agouti-related peptide (AGRP), underpinned this phenotype. Blocking the action of FBPase returned food intake and body weight to those of the negative littermates. Our study is the first to identify liver FBPase as a previously unknown regulator of appetite and adiposity. Importantly, this work recognizes the liver as an important organ in appetite and body weight regulation. This commentary will provide further insight and expand on this novel concept that the liver does in fact play an important role in adiposity.
Collapse
|
50
|
|