1
|
Chu DT, Thi HV, Bui NL, Le NH. The effects of a diet with high fat content from lard on the health and adipose-markers' mRNA expression in mice. Sci Prog 2024; 107:368504241269431. [PMID: 39090965 PMCID: PMC11297511 DOI: 10.1177/00368504241269431] [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] [Indexed: 08/04/2024]
Abstract
Pork is one type of the most frequently consumed meat with about 30% globally. Thus, the questions regarding to the health effects of diet with high fat content from lard are raised. Here, we developed a model of mice fed with high fat (HF) from lard to investigate and have more insights on the effects of long-time feeding with HF on health. The results showed that 66 days on HF induced a significant gain in the body weight of mice, and this weight gain was associated to the deposits in the white fat, but not brown fat. The glucose tolerance, not insulin resistance, in mice was decreased by the HF diet, and this was accompanied with significantly higher blood levels of total cholesterol and triglycerides. Furthermore, the weight gains in mice fed with HF seemed to link to increased mRNA levels of adipose biomarkers in lipogenesis, including Acly and Acaca genes, in white fat tissues. Thus, our study shows that a diet with high fat from lard induced the increase in body weight, white fat depots' expansion, disruption of glucose tolerance, blood dyslipidemia, and seemed to start affecting the mRNA expression of some adipose biomarkers in a murine model.
Collapse
Affiliation(s)
- Dinh-Toi Chu
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam
- Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam
| | - Hue Vu Thi
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam
- Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam
| | - Nhat-Le Bui
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam
- Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam
| | - Ngoc-Hoan Le
- Faculty of Biology, Hanoi National University of Education, Hanoi, Vietnam
| |
Collapse
|
2
|
Gómez-Vilarrubla A, Mas-Parés B, Carreras-Badosa G, Bonmatí-Santané A, Martínez-Calcerrada JM, Niubó-Pallàs M, de Zegher F, Ibáñez L, López-Bermejo A, Bassols J. DNA Methylation Signatures in Paired Placenta and Umbilical Cord Samples: Relationship with Maternal Pregestational Body Mass Index and Offspring Metabolic Outcomes. Biomedicines 2024; 12:301. [PMID: 38397903 PMCID: PMC10886657 DOI: 10.3390/biomedicines12020301] [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: 11/28/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
An epigenomic approach was used to study the impact of maternal pregestational body mass index (BMI) on the placenta and umbilical cord methylomes and their potential effect on the offspring's metabolic phenotype. DNA methylome was assessed in 24 paired placenta and umbilical cord samples. The differentially methylated CpGs associated with maternal pregestational BMI were identified and the metabolic pathways and the potentially related diseases affected by their annotated genes were determined. Two top differentially methylated CpGs were studied in 90 additional samples and the relationship with the offspring's metabolic phenotype was determined. The results showed that maternal pregestational BMI is associated with the methylation of genes involved in endocrine and developmental pathways with potential effects on type 2 diabetes and obesity. The methylation and expression of HADHA and SLC2A8 genes in placenta and umbilical cord were related to several metabolic parameters in the offspring at 6 years (weight SDS, height SDS, BMI SDS, Δ BW-BMI SDS, FM SDS, waist, SBP, TG, HOMA-IR, perirenal fat; all p < 0.05). Our data suggest that epigenetic analysis in placenta and umbilical cord may be useful for identifying individual vulnerability to later metabolic diseases.
Collapse
Affiliation(s)
- Ariadna Gómez-Vilarrubla
- Maternal-Fetal Metabolic Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190 Salt, Spain
| | - Berta Mas-Parés
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190 Salt, Spain
| | - Gemma Carreras-Badosa
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190 Salt, Spain
| | | | | | - Maria Niubó-Pallàs
- Maternal-Fetal Metabolic Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190 Salt, Spain
| | - Francis de Zegher
- Department of Development & Regeneration, University of Leuven, 3000 Leuven, Belgium;
| | - Lourdes Ibáñez
- Endocrinology, Pediatric Research Institute, Sant Joan de Déu Children’s Hospital, 08950 Esplugues de Llobregat, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Health Institute Carlos III (ISCIII), 28029 Madrid, Spain
| | - Abel López-Bermejo
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190 Salt, Spain
- Department of Pediatrics, Dr. Josep Trueta Hospital, 17007 Girona, Spain
| | - Judit Bassols
- Maternal-Fetal Metabolic Research Group, Girona Institute for Biomedical Research (IDIBGI), 17190 Salt, Spain
| |
Collapse
|
3
|
Poudel A, Gachumi G, Paterson PG, El-Aneed A, Badea I. Liposomal Phytosterols as LDL-Cholesterol-Lowering Agents in Diet-Induced Hyperlipidemia. Mol Pharm 2023; 20:4443-4452. [PMID: 37492942 DOI: 10.1021/acs.molpharmaceut.2c01072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
The high blood level of low-density lipoprotein cholesterol (LDL-C) is a primary risk factor for cardiovascular disease. Plant sterols, known as phytosterols (PSs), can reduce LDL-C in a range of 8-14%. The extent of LDL-C reduction depends on its formulation. Encapsulation into liposomes is one formulation strategy to enhance the efficiency of PSs. PSs (campesterol, stigmasterol, and β-sitosterol) have frequently been assessed alone or in combination for their LDL-C-lowering ability. However, one naturally abundant PS, brassicasterol, has not yet been tested for its efficacy. We have previously developed a novel liposomal formulation containing the PS mixture present naturally in canola that is composed of brassicasterol, campesterol, and β-sitosterol. In this work, the efficacy of our novel liposomal PS formulation that includes brassicasterol was assessed in a hamster model. Animals were divided into five groups: (i) liposomal PS in orange juice, (ii) liposomal PS in water, (iii) marketed PS in orange juice, (iv) control orange juice, and (v) control water. The animals were fed a high-fat, cholesterol-supplemented (0.5%) diet to induce hypercholesterolemia. The treatment was administered orally once daily for 4 weeks. Fasting blood samples were collected at baseline, week 2, and week 4. The extent of the reduction of total cholesterol, LDL-C, high-density lipoprotein cholesterol (HDL-C), and triglycerides was compared among the groups. Liposomal PSs in both orange juice and water significantly reduced LDL-C compared to their controls. Furthermore, the liposomal PS was as effective as a marketed PS-containing product in reducing LDL-C. Liposomal PSs in both orange juice and water showed similar efficacy in LDL-C reduction, highlighting that these vehicles/food matrices do not affect the efficacy of PSs. The liposomal formulation of a natural PS mixture extracted from canola oil, with brassicasterol as a major component, exhibited a significant LDL-C reduction in a hamster model.
Collapse
Affiliation(s)
- Asmita Poudel
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon S7N 5E5, Saskatchewan, Canada
| | - George Gachumi
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon S7N 5E5, Saskatchewan, Canada
| | - Phyllis G Paterson
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon S7N 5E5, Saskatchewan, Canada
| | - Anas El-Aneed
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon S7N 5E5, Saskatchewan, Canada
| | - Ildiko Badea
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon S7N 5E5, Saskatchewan, Canada
| |
Collapse
|
4
|
Zuo Q, Zhang G, He L, Ma S, Ma H, Zhai J, Wang Z, Zhang T, Wang Y, Guo Y. Canagliflozin Attenuates Hepatic Steatosis and Atherosclerosis Progression in Western Diet-Fed ApoE-Knockout Mice. Drug Des Devel Ther 2022; 16:4161-4177. [PMID: 36510490 PMCID: PMC9741490 DOI: 10.2147/dddt.s388823] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Purpose To investigate the effect of canagliflozin (20 mg/kg) on hepatic steatosis and atherosclerosis, and further to explore its possible mechanism. Methods Blood glucose, blood lipid, oxidative stress response and inflammatory cytokines were examined by intraperitoneal glucose tolerance test and ELISA assay. HE and Oil Red O staining were used to estimate the extent of hepatic steatosis and atherosclerosis. RNA-seq and qRT-PCR were used to further investigate the potential mechanism. The effects of canagliflozin on autophagy were detected using transmission electron microscopy and Western blotting. The endothelial function-related markers were determined by qRT-PCR. Results Canagliflozin notably alleviated the elevation in blood glucose and insulin resistance in western diet-fed ApoE-/- mice. In ApoE-/-+Cana group, ApoE-/- mice had lower levels of TG, TC, LDL-C, TNF-α, IL-6, IL-1β, and MCP-1. HE and Oil Red O staining presented that canagliflozin restrained the atherosclerotic plaque development and lipid accumulation. RNA-seq showed that 87 DEGs were relevant to improvement of hepatic steatosis and atherosclerosis by canagliflozin. Among them, CPS1, ASS1, ASL, ARG1, MATLA, GLS2, GOT1, SREBP1, Plin5, Retreg1, and C/EBPβ were verified. KEGG enrichment analysis indicated that DEGs were mainly involved in amino acid metabolism. Besides, we observed that canagliflozin reduced the contents of aspartic acid and citrulline in liver. Western blotting showed that ASS1 and p-AMPK/AMPK was remarkably elevated after administration of canagliflozin. Correspondingly, canagliflozin down-regulated SREBP1, FAS, ACC1, HMGCR, p-mTOR/m-TOR, p-ULK1/ULK1 and p62, but up-regulated CPT1, Beclin 1 and LC3 II/LC3I. TEM showed that canagliflozin reduced the number of lipid droplets and increased the autophagosomes. Moreover, we found that canagliflozin elevated the aortic endothelial function-associated markers including ASS1, ASL and eNOS. Conclusion Canagliflozin may attenuate hepatic steatosis by improving lipid metabolism, enhancing autophagy, and reducing inflammatory response through ASS1/AMPK pathway. Besides, canagliflozin further effectively improves the aortic endothelial function, thereby suppressing atherosclerosis development.
Collapse
Affiliation(s)
- Qingjuan Zuo
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, People’s Republic of China,Department of Geriatric Cardiology, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Guorui Zhang
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, People’s Republic of China,Department of Cardiology, the Third Hospital of Shijiazhuang City Affiliated to Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Lili He
- Department of Geriatric Cardiology, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Sai Ma
- Department of Internal Medicine, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Huijuan Ma
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Jianlong Zhai
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Zhongli Wang
- Department of Physical Examination Center, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Tingting Zhang
- Department of Geriatric Cardiology, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Yan Wang
- Department of Geriatric Cardiology, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Yifang Guo
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, People’s Republic of China,Department of Geriatric Cardiology, Hebei General Hospital, Shijiazhuang, People’s Republic of China,Correspondence: Yifang Guo, Department of Geriatric Cardiology, Hebei General Hospital, No. 348, Heping West Road, Xinhua District, Shijiazhuang, Hebei, 050051, People’s Republic of China, Tel +86-15100189182, Email
| |
Collapse
|
5
|
Yang MH, Li WY, Wu CF, Lee YC, Chen AYN, Tyan YC, Chen YMA. Reversal of High-Fat Diet-Induced Non-Alcoholic Fatty Liver Disease by Metformin Combined with PGG, an Inducer of Glycine N-Methyltransferase. Int J Mol Sci 2022; 23:ijms231710072. [PMID: 36077467 PMCID: PMC9456083 DOI: 10.3390/ijms231710072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/27/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a major cause of liver-related morbidities and mortality, and no effective drug treatment currently exists. We aimed to develop a novel treatment strategy to induce the expression of glycine N-methyltransferase (GNMT), which is an important enzyme regulating S-adenosylmethionine metabolism whose expression is downregulated in patients with NAFLD. Because 1,2,3,4,6-pentagalloyl glucose (PGG) is a GNMT inducer, and metformin was shown to upregulate liver mitochondrial GNMT protein expression, the effect of PGG and metformin was evaluated. Biochemical analysis, histopathological examination, immunohistochemical staining, reverse transcription-quantitative PCR (RT-qPCR), Western blotting (WB), proteomic analysis and Seahorse XF Cell Mito Stress Test were performed. The high-fat diet (HFD)-induced NAFLD mice were treated with PGG and metformin. Combination of PGG and metformin nearly completely reversed weight gain, elevation of serum aminotransferases, and hepatic steatosis and steatohepatitis. In addition, the downregulated GNMT expression in liver tissues of HFD-induced NAFLD mice was restored. The GNMT expression was further confirmed by RT-qPCR and WB analysis using both in vitro and in vivo systems. In addition, PGG treatment was shown to increase oxygen consumption rate (OCR) maximum capacity in a dose-dependent manner, and was capable of rescuing the suppression of mitochondrial OCR induced by metformin. Proteomic analysis identified increased expression of glutathione S-transferase mu 4 (GSTM4), heat shock protein 72 (HSP72), pyruvate carboxylase (PYC) and 40S ribosomal protein S28 (RS28) in the metformin plus PGG treatment group. Our findings show that GNMT expression plays an important role in the pathogenesis of NAFLD, and combination of an inducer of GNMT and metformin can be of therapeutic potential for patients with NAFLD.
Collapse
Affiliation(s)
- Ming-Hui Yang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
- Center of General Education, Shu-Zen Junior College of Medicine and Management, Kaohsiung 821, Taiwan
| | - Wei-You Li
- Laboratory of Important Infectious Diseases and Cancer, Graduate Institute of Biomedical and Pharmacological Science, School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Ching-Fen Wu
- Department of Veterinary Medicine, National Chiayi University, Chiayi City 600, Taiwan
| | - Yi-Ching Lee
- Laboratory of Important Infectious Diseases and Cancer, Graduate Institute of Biomedical and Pharmacological Science, School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Allan Yi-Nan Chen
- School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Yu-Chang Tyan
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Nuclear Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
- School of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Correspondence: (Y.-C.T.); (Y.-M.A.C.)
| | - Yi-Ming Arthur Chen
- Laboratory of Important Infectious Diseases and Cancer, Graduate Institute of Biomedical and Pharmacological Science, School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County 350, Taiwan
- Correspondence: (Y.-C.T.); (Y.-M.A.C.)
| |
Collapse
|
6
|
Lactucin & Lactucopicrin ameliorates FFA-induced steatosis in HepG2 cells via modulating lipid metabolism. J Pharmacol Sci 2022; 150:110-122. [DOI: 10.1016/j.jphs.2022.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/09/2022] [Accepted: 07/28/2022] [Indexed: 11/19/2022] Open
|
7
|
Lotus seed resistant starch ameliorates high-fat diet induced hyperlipidemia by fatty acid degradation and glycerolipid metabolism pathways in mouse liver. Int J Biol Macromol 2022; 215:79-91. [PMID: 35718147 DOI: 10.1016/j.ijbiomac.2022.06.077] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/02/2022] [Accepted: 06/11/2022] [Indexed: 11/23/2022]
Abstract
We investigated the potential efficacy and underlying mechanisms of Lotus seed Resistant Starch (LRS) for regulating hyperlipidemia in mice fed a High-fat Diet (HFD). Mouse were fed a normal diet (Normal Control group, NC group), HFD alone (MC group), HFD plus lovastatin (PC group), or HFD with low/medium/high LRS (LLRS, MLRS, and HLRS groups, respectively) for 4 weeks. LRS supplementation significantly decreased body weight and significantly reduced serum levels of total cholesterol, triglycerides, low-density lipoprotein cholesterol, and high-density lipopro-tein cholesterol compared with the MC group. LRS also significantly alleviated hepatic steatosis, especially in the MLRS group, which also showed a significantly reduced visceral fat index. LLRS supplementation significantly regulated genes associated with glycerolipid metabolism and steroid hormone biosynthesis (Lpin1 and Ugt2b38), MLRS significantly regulated genes related to fatty acid degradation, fatty acid elongation, and glycerolipid metabolism (Lpin1, Hadha, Aldh3a2, and Acox1), whereas HLRS significantly regulated genes related to fatty acid elongation and glycerolipid metabolism (Lpin1, Elovl3, Elovol5, and Agpat3). The fatty acid-degradation pathway regulated by MLRS thus exerts better control of serum lipid levels, body weight, visceral fat index, and liver steatosis in mice compared with LLRS- and HLRS-regulated pathways.
Collapse
|
8
|
Betaine Supplementation Causes an Increase in Fatty Acid Oxidation and Carbohydrate Metabolism in Livers of Mice Fed a High-Fat Diet: A Proteomic Analysis. Foods 2022; 11:foods11060881. [PMID: 35327303 PMCID: PMC8949908 DOI: 10.3390/foods11060881] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/04/2022] [Accepted: 03/15/2022] [Indexed: 11/16/2022] Open
Abstract
Betaine, a common methyl donor whose methylation is involved in the biosynthesis of carnitine and phospholipids in animals, serves as food and animal feed additive. The present study used liquid chromatography-mass spectrometry (LC-MS) to analyze the liver protein profile of mice on a high fat (HF) diet to investigate the mechanism by which betaine affects hepatic metabolism. Although betaine supplementation had no significant effect on body weight, a total of 103 differentially expressed proteins were identified between HF diet + 1% betaine group (HFB) and HF diet group by LC-MS (fold change > 2, p < 0.05). The addition of 1% betaine had a significant enhancement of the expression of enzymes related to fatty acid oxidation metabolism, such as hydroxyacyl-Coenzyme A dehydrogenase (HADHA), enoyl Coenzyme A hydratase 1 (ECHS1) (p < 0.05) etc., and the expression of apolipoprotein A-II (APOA2) protein was significantly reduced (p < 0.01). Meanwhile, the protein expression of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and succinate-CoA ligase (SUCLG1) were highly significant (p < 0.01). Pathway enrichment using the Kyoto Encyclopedia of Genes and Genomes (KEGG) revealed that the functions of differential proteins involved fatty acid catabolism, carbohydrate metabolism, tricarboxylic acid cycle (TCA) and peroxisome proliferator-activated receptor alpha (PPARα) signaling pathway. Protein−protein interaction (PPI) analysis discovered that acetyl-Coenzyme A acetyltransferase 1 (ACAT1), HADHA and ECHS1 were central hubs of hepatic proteomic changes in the HFB group of mice. Betaine alleviates hepatic lipid accumulation by enhancing fatty acid oxidation and accelerating the TCA cycle and glycolytic process in the liver of mice on an HF diet.
Collapse
|
9
|
Couchet M, Breuillard C, Corne C, Rendu J, Morio B, Schlattner U, Moinard C. Ornithine Transcarbamylase - From Structure to Metabolism: An Update. Front Physiol 2021; 12:748249. [PMID: 34658931 PMCID: PMC8517447 DOI: 10.3389/fphys.2021.748249] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/07/2021] [Indexed: 12/30/2022] Open
Abstract
Ornithine transcarbamylase (OTC; EC 2.1.3.3) is a ubiquitous enzyme found in almost all organisms, including vertebrates, microorganisms, and plants. Anabolic, mostly trimeric OTCs catalyze the production of L-citrulline from L-ornithine which is a part of the urea cycle. In eukaryotes, such OTC localizes to the mitochondrial matrix, partially bound to the mitochondrial inner membrane and part of channeling multi-enzyme assemblies. In mammals, mainly two organs express OTC: the liver, where it is an integral part of the urea cycle, and the intestine, where it synthesizes citrulline for export and plays a major role in amino acid homeostasis, particularly of L-glutamine and L-arginine. Here, we give an overview on OTC genes and proteins, their tissue distribution, regulation, and physiological function, emphasizing the importance of OTC and urea cycle enzymes for metabolic regulation in human health and disease. Finally, we summarize the current knowledge of OTC deficiency, a rare X-linked human genetic disorder, and its emerging role in various chronic pathologies.
Collapse
Affiliation(s)
- Morgane Couchet
- Université Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France
| | - Charlotte Breuillard
- Université Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France
| | | | - John Rendu
- Centre Hospitalier Université Grenoble Alpes, Grenoble, France
| | - Béatrice Morio
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Lyon, France
| | - Uwe Schlattner
- Université Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France.,Institut Universitaire de France, Paris, France
| | - Christophe Moinard
- Université Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France
| |
Collapse
|
10
|
Ha NT, Lee CH. Roles of Farnesyl-Diphosphate Farnesyltransferase 1 in Tumour and Tumour Microenvironments. Cells 2020; 9:cells9112352. [PMID: 33113804 PMCID: PMC7693003 DOI: 10.3390/cells9112352] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/23/2020] [Accepted: 10/24/2020] [Indexed: 12/14/2022] Open
Abstract
Farnesyl-diphosphate farnesyltransferase 1 (FDFT1, squalene synthase), a membrane-associated enzyme, synthesizes squalene via condensation of two molecules of farnesyl pyrophosphate. Accumulating evidence has noted that FDFT1 plays a critical role in cancer, particularly in metabolic reprogramming, cell proliferation, and invasion. Based on these advances in our knowledge, FDFT1 could be a potential target for cancer treatment. This review focuses on the contribution of FDFT1 to the hallmarks of cancer, and further, we discuss the applicability of FDFT1 as a cancer prognostic marker and target for anticancer therapy.
Collapse
|
11
|
Whittaker K, Burgess R, Jones V, Yang Y, Zhou W, Luo S, Wilson J, Huang R. Quantitative proteomic analyses in blood: A window to human health and disease. J Leukoc Biol 2019; 106:759-775. [PMID: 31329329 DOI: 10.1002/jlb.mr1118-440r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/11/2019] [Accepted: 06/24/2019] [Indexed: 12/13/2022] Open
Affiliation(s)
| | | | | | | | | | - Shuhong Luo
- RayBiotech Life Norcross Georgia USA
- RayBiotech Life Guangzhou Guangdong China
- South China Biochip Research Center Guangzhou Guangdong China
| | | | - Ruo‐Pan Huang
- RayBiotech Life Norcross Georgia USA
- RayBiotech Life Guangzhou Guangdong China
- South China Biochip Research Center Guangzhou Guangdong China
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityGuangzhou Medical University Guangzhou China
- Guangdong Provincial Hospital of Chinese Medicine Guangzhou China
| |
Collapse
|
12
|
Cholesterol-Lowering and Liver-Protective Effects of Cooked and Germinated Mung Beans ( Vigna radiata L.). Nutrients 2018; 10:nu10070821. [PMID: 29949855 PMCID: PMC6073478 DOI: 10.3390/nu10070821] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 06/19/2018] [Accepted: 06/25/2018] [Indexed: 12/04/2022] Open
Abstract
We investigated the hypocholesterolemic and liver-protective effects of cooked and germinated whole mung beans. Hamsters were fed for 28 days on diets rich in saturated fatty acids and cholesterol, differing only in protein source (20%): casein, cooked whole mung bean, and germinated mung bean. After 28 days, we found reduced plasma concentrations of total cholesterol and non-HDL cholesterol, increased faecal cholesterol excretion, and reduced levels of asparagine aminotransferase and alanine aminotransferase enzymes in the liver. Reduction in hepatic lipid deposition was observed between each of the mung bean groups relative to the casein group. In addition, the animals of the geminated mung bean group showed a lack of inflammatory infiltrate and better vascularisation of the hepatic tissue. Results from this study show significant hypocholesterolemic and liver-protective properties of the mung bean, which are further enhanced after germination.
Collapse
|
13
|
Diez-Fernandez C, Rüfenacht V, Gemperle C, Fingerhut R, Häberle J. Mutations and common variants in the human arginase 1 (ARG1
) gene: Impact on patients, diagnostics, and protein structure considerations. Hum Mutat 2018; 39:1029-1050. [DOI: 10.1002/humu.23545] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/20/2018] [Accepted: 04/25/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Carmen Diez-Fernandez
- University Children's Hospital Zurich; Division of Metabolism and Children's Research Center; Zurich Switzerland
| | - Véronique Rüfenacht
- University Children's Hospital Zurich; Division of Metabolism and Children's Research Center; Zurich Switzerland
| | - Corinne Gemperle
- University Children's Hospital Zurich; Division of Metabolism and Children's Research Center; Zurich Switzerland
| | - Ralph Fingerhut
- University Children's Hospital Zurich; Division of Metabolism and Children's Research Center; Zurich Switzerland
| | - Johannes Häberle
- University Children's Hospital Zurich; Division of Metabolism and Children's Research Center; Zurich Switzerland
| |
Collapse
|
14
|
Wan W, Li H, Xiang J, Yi F, Xu L, Jiang B, Xiao P. Aqueous Extract of Black Maca Prevents Metabolism Disorder via Regulating the Glycolysis/Gluconeogenesis-TCA Cycle and PPARα Signaling Activation in Golden Hamsters Fed a High-Fat, High-Fructose Diet. Front Pharmacol 2018; 9:333. [PMID: 29681858 PMCID: PMC5897445 DOI: 10.3389/fphar.2018.00333] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 03/22/2018] [Indexed: 12/19/2022] Open
Abstract
Maca (Lepidium meyenii Walpers) has been used as a dietary supplement and ethnomedicine for centuries. Recently, maca has become a high profile functional food worldwide because of its multiple biological activities. This study is the first explorative research to investigate the prevention and amelioration capacity of the aqueous extract of black maca (AEM) on high-fat, high-fructose diet (HFD)-induced metabolism disorder in golden hamsters and to identify the potential mechanisms involved in these effects. For 20 weeks, 6-week-old male golden hamsters were fed the following respective diets: (1) a standard diet, (2) HFD, (3) HFD supplemented with metformin, or (4) HFD supplemented with three doses of AEM (300, 600, or 1,200 mg/kg). After 20 weeks, the golden hamsters that received daily AEM supplementation presented with the beneficial effects of improved hyperlipidemia, hyperinsulinemia, insulin resistance, and hepatic steatosis in vivo. Based on the hepatic metabolomic analysis results, alterations in metabolites associated with pathological changes were examined. A total of 194 identified metabolites were mapped to 46 relative metabolic pathways, including those of energy metabolism. In addition, via in silico profiling for secondary maca metabolites by a joint pharmacophore- and structure-based approach, a compound-target-disease network was established. The results revealed that 32 bioactive compounds in maca targeted 16 proteins involved in metabolism disorder. Considering the combined metabolomics and virtual screening results, we employed quantitative real-time PCR assays to verify the gene expression of key enzymes in the relevant pathways. AEM promoted glycolysis and inhibited gluconeogenesis via regulating the expression of key genes such as Gck and Pfkm. Moreover, AEM upregulated tricarboxylic acid (TCA) cycle flux by changing the concentrations of intermediates and increasing the mRNA levels of Aco2, Fh, and Mdh2. In addition, the lipid-lowering effects of AEM in boththe serum and liver may be partly related to PPARα signaling activation, including enhanced fatty acid β-oxidation and lipogenesis pathway inhibition. Together, our data demonstrated that AEM intervention significantly improved lipid and glucose metabolism disorder by regulating the glycolysis/gluconeogenesis-TCA cycle and by modulating gene expression levels involved in the PPARα signaling pathway.
Collapse
Affiliation(s)
- Wenting Wan
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Hongxiang Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Jiamei Xiang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Fan Yi
- School of Sciences/Key Laboratory of Cosmetic, China National Light Industry, Beijing Technology and Business University, Beijing, China
| | - Lijia Xu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Baoping Jiang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Peigen Xiao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| |
Collapse
|
15
|
Sridharan GV, D'Alessandro M, Bale SS, Bhagat V, Gagnon H, Asara JM, Uygun K, Yarmush ML, Saeidi N. Multi-omic network-based interrogation of rat liver metabolism following gastric bypass surgery featuring SWATH proteomics. TECHNOLOGY 2017; 5:139-184. [PMID: 29780857 PMCID: PMC5956888 DOI: 10.1142/s233954781750008x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Morbidly obese patients often elect for Roux-en-Y gastric bypass (RYGB), a form of bariatric surgery that triggers a remarkable 30% reduction in excess body weight and reversal of insulin resistance for those who are type II diabetic. A more complete understanding of the underlying molecular mechanisms that drive the complex metabolic reprogramming post-RYGB could lead to innovative non-invasive therapeutics that mimic the beneficial effects of the surgery, namely weight loss, achievement of glycemic control, or reversal of non-alcoholic steatohepatitis (NASH). To facilitate these discoveries, we hereby demonstrate the first multi-omic interrogation of a rodent RYGB model to reveal tissue-specific pathway modules implicated in the control of body weight regulation and energy homeostasis. In this study, we focus on and evaluate liver metabolism three months following RYGB in rats using both SWATH proteomics, a burgeoning label free approach using high resolution mass spectrometry to quantify protein levels in biological samples, as well as MRM metabolomics. The SWATH analysis enabled the quantification of 1378 proteins in liver tissue extracts, of which we report the significant down-regulation of Thrsp and Acot13 in RYGB as putative targets of lipid metabolism for weight loss. Furthermore, we develop a computational graph-based metabolic network module detection algorithm for the discovery of non-canonical pathways, or sub-networks, enriched with significantly elevated or depleted metabolites and proteins in RYGB-treated rat livers. The analysis revealed a network connection between the depleted protein Baat and the depleted metabolite taurine, corroborating the clinical observation that taurine-conjugated bile acid levels are perturbed post-RYGB.
Collapse
Affiliation(s)
- Gautham Vivek Sridharan
- Center for Engineering in Medicine, Harvard Medical School - Massachusetts General Hospital, 51 Blossom Street, Boston, MA 02114, USA
| | - Matthew D'Alessandro
- Center for Engineering in Medicine, Harvard Medical School - Massachusetts General Hospital, 51 Blossom Street, Boston, MA 02114, USA
| | - Shyam Sundhar Bale
- Center for Engineering in Medicine, Harvard Medical School - Massachusetts General Hospital, 51 Blossom Street, Boston, MA 02114, USA
| | - Vicky Bhagat
- Warren Alpert Medical School of Brown University, 222 Richmond St., Providence, RI 02903, USA
| | - Hugo Gagnon
- Phenoswitch Bioscience, 3001 12e Avenue N, Sherbrooke, QC J1H 5N4, Canada
| | - John M Asara
- Beth Israel Deaconness Medical Center, 3 Blackfan Circle Rm 425, Boston, MA 02115, USA
| | - Korkut Uygun
- Center for Engineering in Medicine, Harvard Medical School - Massachusetts General Hospital, 51 Blossom Street, Boston, MA 02114, USA
| | - Martin L Yarmush
- Center for Engineering in Medicine, Harvard Medical School - Massachusetts General Hospital, 51 Blossom Street, Boston, MA 02114, USA
| | - Nima Saeidi
- Center for Engineering in Medicine, Harvard Medical School - Massachusetts General Hospital, 51 Blossom Street, Boston, MA 02114, USA
| |
Collapse
|
16
|
Janssens S, Ciapaite J, Wolters JC, van Riel NA, Nicolay K, Prompers JJ. An In Vivo Magnetic Resonance Spectroscopy Study of the Effects of Caloric and Non-Caloric Sweeteners on Liver Lipid Metabolism in Rats. Nutrients 2017; 9:nu9050476. [PMID: 28489050 PMCID: PMC5452206 DOI: 10.3390/nu9050476] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/26/2017] [Accepted: 05/03/2017] [Indexed: 12/14/2022] Open
Abstract
We aimed to elucidate the effects of caloric and non-caloric sweeteners on liver lipid metabolism in rats using in vivo magnetic resonance spectroscopy (MRS) and to determine their roles in the development of liver steatosis. Wistar rats received normal chow and either normal drinking water, or solutions containing 13% (w/v) glucose, 13% fructose, or 0.4% aspartame. After 7 weeks, in vivo hepatic dietary lipid uptake and de novo lipogenesis were assessed with proton-observed, carbon-13-edited MRS combined with 13C-labeled lipids and 13C-labeled glucose, respectively. The molecular basis of alterations in hepatic liver metabolism was analyzed in detail ex vivo using immunoblotting and targeted quantitative proteomics. Both glucose and fructose feeding increased adiposity, but only fructose induced hepatic lipid accumulation. In vivo MRS showed that this was not caused by increased hepatic uptake of dietary lipids, but could be attributed to an increase in de novo lipogenesis. Stimulation of lipogenesis by fructose was confirmed by a strong upregulation of lipogenic enzymes, which was more potent than with glucose. The non-caloric sweetener aspartame did not significantly affect liver lipid content or metabolism. In conclusion, liquid fructose more severely affected liver lipid metabolism in rats than glucose, while aspartame had no effect.
Collapse
Affiliation(s)
- Sharon Janssens
- Biomedical Nuclear Magnetic Resonance (NMR), Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands.
| | - Jolita Ciapaite
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
- Systems Biology Centre for Energy Metabolism and Ageing, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| | - Justina C Wolters
- Systems Biology Centre for Energy Metabolism and Ageing, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
- Department of Pharmacy, Analytical Biochemistry, University of Groningen, Antonius Deusinglaan, 9713 AV Groningen, The Netherlands.
| | - Natal A van Riel
- Computational Biology, Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands.
| | - Klaas Nicolay
- Biomedical Nuclear Magnetic Resonance (NMR), Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands.
| | - Jeanine J Prompers
- Biomedical Nuclear Magnetic Resonance (NMR), Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands.
| |
Collapse
|
17
|
Park HM, Park KT, Park EC, Kim SI, Choi MS, Liu KH, Lee CH. Mass Spectrometry-Based Metabolomic and Lipidomic Analyses of the Effects of Dietary Platycodon grandiflorum on Liver and Serum of Obese Mice under a High-Fat Diet. Nutrients 2017; 9:nu9010071. [PMID: 28106735 PMCID: PMC5295115 DOI: 10.3390/nu9010071] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 12/27/2016] [Accepted: 01/07/2017] [Indexed: 12/23/2022] Open
Abstract
We aimed to identify metabolites involved in the anti-obesity effects of Platycodon grandiflorum (PG) in high-fat diet (HFD)-fed mice using mass spectrometry (MS)-based metabolomic techniques. C57BL/6J mice were divided into four groups: normal diet (ND)-fed mice, HFD-fed mice, HFD with 1% PG extract-fed mice (HPGL), and HFD with 5% PG extract-fed mice (HPGH). After 8 weeks, the HFD group gained more weight than the ND group, while dietary 5% PG extract attenuated this change. The partial least squares discriminant analysis (PLS-DA) score plots showed a clear distinction between experimental groups in serum and liver markers. We also identified 10 and 32 metabolites in the serum and liver, respectively, as potential biomarkers that could explain the effect of high-dose PG added to HFD-fed mice, which were strongly involved in amino acid metabolism (glycine, serine, threonine, methionine, glutamate, phenylalanine, ornithine, lysine, and tyrosine), TCA cycle (fumarate and succinate), lipid metabolism (linoleic and oleic acid methyl esters, oleamide, and cholesterol), purine/pyrimidine metabolism (uracil and hypoxanthine), carbohydrate metabolism (maltose), and glycerophospholipid metabolism (phosphatidylcholines, phosphatidylethanolamines, lysophosphatidylcholines, and lysophosphatidylethanolamines). We suggest that further studies on these metabolites could help us gain a better understanding of both HFD-induced obesity and the effects of PG.
Collapse
Affiliation(s)
- Hye Min Park
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| | - Kab-Tae Park
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea.
| | - Edmond Changkyun Park
- Division of Life Science, Korea Basic Science Institute, Daejeon 34133, Korea.
- Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Korea.
| | - Seung Ii Kim
- Division of Life Science, Korea Basic Science Institute, Daejeon 34133, Korea.
- Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Korea.
| | - Myung Sook Choi
- Department of Food Science and Nutrition, Kyungpook National University, Daegu 41566, Korea.
| | - Kwang-Hyeon Liu
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea.
| | - Choong Hwan Lee
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| |
Collapse
|
18
|
Somvanshi PR, Patel AK, Bhartiya S, Venkatesh KV. Influence of plasma macronutrient levels on hepatic metabolism: role of regulatory networks in homeostasis and disease states. RSC Adv 2016. [DOI: 10.1039/c5ra18128c] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Multilevel regulations by metabolic, signaling and transcription pathways form a complex network that works to provide robust metabolic regulation in the liver. This analysis indicates that dietary perturbations in these networks can lead to insulin resistance.
Collapse
Affiliation(s)
- Pramod R. Somvanshi
- Biosystems Engineering Lab
- Department of Chemical Engineering
- Indian Institute of Technology Bombay
- Mumbai
- India 400076
| | - Anilkumar K. Patel
- Biosystems Engineering Lab
- Department of Chemical Engineering
- Indian Institute of Technology Bombay
- Mumbai
- India 400076
| | - Sharad Bhartiya
- Control Systems Engineering Lab
- Department of Chemical Engineering
- Indian Institute of Technology Bombay
- Mumbai
- India 400076
| | - K. V. Venkatesh
- Biosystems Engineering Lab
- Department of Chemical Engineering
- Indian Institute of Technology Bombay
- Mumbai
- India 400076
| |
Collapse
|
19
|
Pedersen BA, Wang W, Taylor JF, Khattab OS, Chen YH, Edwards RA, Yazdi PG, Wang PH. Hepatic proteomic analysis revealed altered metabolic pathways in insulin resistant Akt1(+/-)/Akt2(-/-) mice. Metabolism 2015; 64:1694-703. [PMID: 26455965 PMCID: PMC4641788 DOI: 10.1016/j.metabol.2015.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 08/19/2015] [Accepted: 09/08/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVE The aim of this study was to identify liver proteome changes in a mouse model of severe insulin resistance and markedly decreased leptin levels. METHODS Two-dimensional differential gel electrophoresis was utilized to identify liver proteome changes in AKT1(+/-)/AKT2(-/-) mice. Proteins with altered levels were identified with tandem mass spectrometry. Ingenuity Pathway Analysis was performed for the interpretation of the biological significance of the observed proteomic changes. RESULTS 11 proteins were identified from 2 biological replicates to be differentially expressed by a ratio of at least 1.3 between age-matched insulin resistant (Akt1(+/-)/Akt2(-/-)) and wild type mice. Albumin and mitochondrial ornithine aminotransferase were detected from multiple spots, which suggest post-translational modifications. Enzymes of the urea cycle were common members of top regulated pathways. CONCLUSION Our results help to unveil the regulation of the liver proteome underlying altered metabolism in an animal model of severe insulin resistance.
Collapse
Affiliation(s)
- Brian A Pedersen
- UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California at Irvine, Irvine, CA 92697, USA
- Department of Medicine, University of California at Irvine, Irvine, CA 92697, USA
| | - Weiwen Wang
- UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California at Irvine, Irvine, CA 92697, USA
- Department of Medicine, University of California at Irvine, Irvine, CA 92697, USA
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami School of Medicine, Miami, FL, 33136
| | - Jared F Taylor
- UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California at Irvine, Irvine, CA 92697, USA
- Department of Medicine, University of California at Irvine, Irvine, CA 92697, USA
| | - Omar S Khattab
- UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California at Irvine, Irvine, CA 92697, USA
| | - Yu-Han Chen
- UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California at Irvine, Irvine, CA 92697, USA
- Department of Physiology & Biophysics, University of California at Irvine, Irvine, CA 92697, USA
| | - Robert A Edwards
- Department of Pathology, University of California at Irvine, Irvine, CA 92697, USA
| | - Puya G Yazdi
- UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California at Irvine, Irvine, CA 92697, USA
- Department of Medicine, University of California at Irvine, Irvine, CA 92697, USA
| | - Ping H Wang
- UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California at Irvine, Irvine, CA 92697, USA
- Department of Medicine, University of California at Irvine, Irvine, CA 92697, USA
- Department of Biological Chemistry, University of California at Irvine, Irvine, CA 92697, USA
- Department of Physiology & Biophysics, University of California at Irvine, Irvine, CA 92697, USA
| |
Collapse
|