1
|
Funes AK, Avena V, Boarelli PV, Monclus MA, Zoppino DF, Saez-Lancellotti TE, Fornes MW. Cholesterol dynamics in rabbit liver: High-fat diet, olive oil, and synergistic dietary effects. Biochem Biophys Res Commun 2024; 733:150675. [PMID: 39284268 DOI: 10.1016/j.bbrc.2024.150675] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/30/2024] [Accepted: 09/06/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND & AIMS Lipid metabolism disorders contribute to a range of human diseases, including liver-related pathologies. Rabbits, highly sensitive to dietary cholesterol, provide a model for understanding the development of liver disorders. Sterol regulatory element-binding protein isoform 2 (SREBP2) crucially regulates intracellular cholesterol pathways. Extra-virgin olive oil (EVOO) has shown reducing cholesterol levels and restoring liver parameters affected by HFD. The aim was to investigate the molecular impact of an HFD and supplemented with EVOO on rabbit liver cholesterol metabolism. APPROACH & RESULTS Male rabbits were assigned to dietary cohorts, including control, acute/chronic HFD, sequential HFD with EVOO, and EVOO. Parameters such as serum lipid profiles, hepatic enzymes, body weight, and molecular analyses. After 6 months of HFD, plasma and hepatic cholesterol increased with decreased SREBP2 and 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGCR) expression. Prolonged HFD increased cholesterol levels, upregulating SREBP2 mRNA and HMGCR protein. Combining this with EVOO lowered cholesterol, increased SREBP2 mRNA, and upregulated low-density lipoprotein receptor (LDLR) expression. HFD-induced metabolic dysfunction-associated fatty liver disease was mitigated by EVOO. In conclusion, the SREBP2 system responds to dietary changes. CONCLUSIONS In rabbits, the SREBP2 system responds to dietary changes. Acute HFD hinders cholesterol synthesis, while prolonged HFD disrupts regulation, causing SREBP2 upregulation. EVOO intake prompts LDLR upregulation, potentially enhancing cholesterol clearance and restoring hepatic alterations.
Collapse
Affiliation(s)
- Abi K Funes
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina; Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina; Laboratorio de Enfermedades Metabólicas (LEM), Universidad Juan Agustín Maza, Mendoza, Argentina
| | - Virginia Avena
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina; Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina; Laboratorio de Enfermedades Metabólicas (LEM), Universidad Juan Agustín Maza, Mendoza, Argentina
| | - Paola V Boarelli
- Laboratorio de Enfermedades Metabólicas (LEM), Universidad Juan Agustín Maza, Mendoza, Argentina
| | - María A Monclus
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina; Instituto de Investigaciones, Facultad de Ciencias Médicas, Universidad del Aconcagua, Mendoza, Argentina
| | - Dario Fernández Zoppino
- Laboratorio de Fisiología Celular y Molecular. Facultad de Ciencias de la Salud. Universidad de Burgos, Burgos, Spain
| | - Tania E Saez-Lancellotti
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina; Instituto de Investigaciones, Facultad de Ciencias Médicas, Universidad del Aconcagua, Mendoza, Argentina; Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA). Universidad de Málaga, Málaga, Spain.
| | - Miguel W Fornes
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
| |
Collapse
|
2
|
Takeuchi Y, Murayama Y, Aita Y, Mehrazad Saber Z, Karkoutly S, Tao D, Katabami K, Ye C, Shikama A, Masuda Y, Izumida Y, Miyamoto T, Matsuzaka T, Kawakami Y, Shimano H, Yahagi N. GR-KLF15 pathway controls hepatic lipogenesis during fasting. FEBS J 2024; 291:259-271. [PMID: 37702262 DOI: 10.1111/febs.16957] [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: 05/28/2023] [Revised: 08/10/2023] [Accepted: 09/11/2023] [Indexed: 09/14/2023]
Abstract
During periods of fasting, the body undergoes a metabolic shift from carbohydrate utilization to the use of fats and ketones as an energy source, as well as the inhibition of de novo lipogenesis and the initiation of gluconeogenesis in the liver. The transcription factor sterol regulatory element-binding protein-1 (SREBP-1), which plays a critical role in the regulation of lipogenesis, is suppressed during fasting, resulting in the suppression of hepatic lipogenesis. We previously demonstrated that the interaction of fasting-induced Kruppel-like factor 15 (KLF15) with liver X receptor serves as the essential mechanism for the nutritional regulation of SREBP-1 expression. However, the underlying mechanisms of KLF15 induction during fasting remain unclear. In this study, we show that the glucocorticoid receptor (GR) regulates the hepatic expression of KLF15 and, subsequently, lipogenesis through the KLF15-SREBP-1 pathway during fasting. KLF15 is necessary for the suppression of SREBP-1 by GR, as demonstrated through experiments using KLF15 knockout mice. Additionally, we show that GR is involved in the fasting response, with heightened binding to the KLF15 enhancer. It has been widely known that the hypothalamic-pituitary-adrenal (HPA) axis regulates the secretion of glucocorticoids and plays a significant role in the metabolic response to undernutrition. These findings demonstrate the importance of the HPA-axis-regulated GR-KLF15 pathway in the regulation of lipid metabolism in the liver during fasting.
Collapse
Affiliation(s)
- Yoshinori Takeuchi
- Nutrigenomics Research Group, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Yuki Murayama
- Nutrigenomics Research Group, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yuichi Aita
- Nutrigenomics Research Group, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Zahra Mehrazad Saber
- Nutrigenomics Research Group, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Samia Karkoutly
- Nutrigenomics Research Group, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Duhan Tao
- Nutrigenomics Research Group, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kyoka Katabami
- Nutrigenomics Research Group, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Chen Ye
- Nutrigenomics Research Group, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Akito Shikama
- Nutrigenomics Research Group, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yukari Masuda
- Nutrigenomics Research Group, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yoshihiko Izumida
- Nutrigenomics Research Group, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Takafumi Miyamoto
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Takashi Matsuzaka
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yasushi Kawakami
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hitoshi Shimano
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Naoya Yahagi
- Nutrigenomics Research Group, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Shimotsuke, Japan
| |
Collapse
|
3
|
Wang S, Zhou Y, Yu R, Ling J, Li B, Yang C, Cheng Z, Qian R, Lin Z, Yu C, Zheng J, Zheng X, Jia Q, Wu W, Wu Q, Chen M, Yuan S, Dong W, Shi Y, Jansen R, Yang C, Hao Y, Yao M, Qin W, Jin H. Loss of hepatic FTCD promotes lipid accumulation and hepatocarcinogenesis by upregulating PPARγ and SREBP2. JHEP Rep 2023; 5:100843. [PMID: 37675273 PMCID: PMC10477690 DOI: 10.1016/j.jhepr.2023.100843] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 06/02/2023] [Accepted: 06/26/2023] [Indexed: 09/08/2023] Open
Abstract
Background & Aims Exploiting key regulators responsible for hepatocarcinogenesis is of great importance for the prevention and treatment of hepatocellular carcinoma (HCC). However, the key players contributing to hepatocarcinogenesis remain poorly understood. We explored the molecular mechanisms underlying the carcinogenesis and progression of HCC for the development of potential new therapeutic targets. Methods The Cancer Genome Atlas-Liver Hepatocellular Carcinoma (TCGA-LIHC) and Genotype-Tissue Expression (GTEx) databases were used to identify genes with enhanced expression in the liver associated with HCC progression. A murine liver-specific Ftcd knockout (Ftcd-LKO) model was generated to investigate the role of formimidoyltransferase cyclodeaminase (FTCD) in HCC. Multi-omics analysis of transcriptomics, metabolomics, and proteomics data were applied to further analyse the molecular effects of FTCD expression on hepatocarcinogenesis. Functional and biochemical studies were performed to determine the significance of loss of FTCD expression and the therapeutic potential of Akt inhibitors in FTCD-deficient cancer cells. Results FTCD is highly expressed in the liver but significantly downregulated in HCC. Patients with HCC and low levels of FTCD exhibited worse prognosis, and patients with liver cirrhosis and low FTCD levels exhibited a notable higher probability of developing HCC. Hepatocyte-specific knockout of FTCD promoted both chronic diethylnitrosamine-induced and spontaneous hepatocarcinogenesis in mice. Multi-omics analysis showed that loss of FTCD affected fatty acid and cholesterol metabolism in hepatocarcinogenesis. Mechanistically, loss of FTCD upregulated peroxisome proliferator-activated receptor (PPAR)γ and sterol regulatory element-binding protein 2 (SREBP2) by regulating the PTEN/Akt/mTOR signalling axis, leading to lipid accumulation and hepatocarcinogenesis. Conclusions Taken together, we identified a FTCD-regulated lipid metabolic mechanism involving PPARγ and SREBP2 signaling in hepatocarcinogenesis and provide a rationale for therapeutically targeting of HCC driven by downregulation of FTCD. Impact and implications Exploiting key molecules responsible for hepatocarcinogenesis is significant for the prevention and treatment of HCC. Herein, we identified formimidoyltransferase cyclodeaminase (FTCD) as the top enhanced gene, which could serve as a predictive and prognostic marker for patients with HCC. We generated and characterised the first Ftcd liver-specific knockout murine model. We found loss of FTCD expression upregulated peroxisome proliferator-activated receptor (PPAR)γ and sterol regulatory element-binding protein 2 (SREBP2) by regulating the PTEN/Akt/mTOR signalling axis, leading to lipid accumulation and hepatocarcinogenesis, and provided a rationale for therapeutic targeting of HCC driven by downregulation of FTCD.
Collapse
Affiliation(s)
- Siying Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yangyang Zhou
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruobing Yu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Ling
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Botai Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Yang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhuoan Cheng
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ruolan Qian
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhang Lin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengtao Yu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaojiao Zheng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingling Zheng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Jia
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Wu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiangxin Wu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengnuo Chen
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengxian Yuan
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Wei Dong
- Department of Pathology, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| | - Yaoping Shi
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Robin Jansen
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Chen Yang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd., Nanjing, China
| | - Yujun Hao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Yao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenxin Qin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haojie Jin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Funes AK, Avena MV, Ibañez J, Simón L, Ituarte L, Colombo R, Roldán A, Conte MI, Monclus MÁ, Boarelli P, Fornés MW, Saez Lancellotti TE. Extra-virgin olive oil ameliorates high-fat diet-induced seminal and testicular disorders by modulating the cholesterol pathway. Andrology 2023; 11:1203-1217. [PMID: 36695747 DOI: 10.1111/andr.13398] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 11/02/2022] [Accepted: 01/02/2023] [Indexed: 01/26/2023]
Abstract
BACKGROUND Rabbits are sensitive to dietary cholesterol and rapidly develop hypercholesterolemia, leading to prominent subfertility. Sterol regulatory element-binding protein isoform 2 drives the intracellular cholesterol pathway in many tissues, including the testicles. Its abnormal regulation could be the mainly responsible for the failure of suppressing cholesterol synthesis in a cholesterol-enriched environment, ultimately leading to testicular and seminal alterations. However, extra-virgin olive oil consumption has beneficial properties that promote lowering of cholesterol levels, including the recovery of seminal parameters altered under a high-fat diet. OBJECTIVES Our goal was to investigate the effects of high-fat diet supplementation with extra-virgin olive oil at the molecular level on rabbit testes, by analyzing sterol regulatory element-binding protein isoform 2 protein and its corresponding downstream effectors. MATERIALS AND METHODS During 12 months, male rabbits were fed a control diet, high-fat diet, or 6-month high-fat diet followed by 6-month high-fat diet plus extra-virgin olive oil. Serum lipids, testosterone levels, bodyweight, and seminal parameters were tested. The mRNA and protein levels of sterol regulatory element-binding protein isoform 2, 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase, and low-density lipoprotein receptor were determined by semi-quantitative polymerase chain reaction and Western blotting techniques. The expression pattern of sterol regulatory element-binding protein isoform 2 protein in the rabbit testicles was studied by indirect immunofluorescence. In addition, testicular cholesterol was detected and quantified by filipin staining and gas chromatography. RESULTS The data showed that the addition of extra-virgin olive oil to high-fat diet reduced testicular cholesterol levels and recovered the expression of sterol regulatory element-binding protein isoform 2, 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase, and low-density lipoprotein receptor initially altered by the high-fat diet. DISCUSSION AND CONCLUSIONS The combination of high-fat diet with extra-virgin olive oil encourages testicular recovery by modifying the expression of the enzymes related to intracellular cholesterol management.
Collapse
Affiliation(s)
- Abi Karenina Funes
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
| | - María Virginia Avena
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
| | - Jorge Ibañez
- Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
| | - Layla Simón
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
| | - Leonor Ituarte
- Área de Física Biológica, Departamento de Morfofisiología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Regina Colombo
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
| | - Adrián Roldán
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Instituto de Investigaciones, Facultad de Ciencias Médicas, Universidad del Aconcagua, Mendoza, Argentina
| | - María Inés Conte
- Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
| | - María Ángeles Monclus
- Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Instituto de Investigaciones, Facultad de Ciencias Médicas, Universidad del Aconcagua, Mendoza, Argentina
| | - Paola Boarelli
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Laboratorio de Enfermedades Metabólicas (LEM), Universidad Juan Agustín Maza, Mendoza, Argentina
| | - Miguel Walter Fornés
- Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
| | - Tania Estefanía Saez Lancellotti
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Instituto de Investigaciones, Facultad de Ciencias Médicas, Universidad del Aconcagua, Mendoza, Argentina
| |
Collapse
|
5
|
Vuu YM, Kadar Shahib A, Rastegar M. The Potential Therapeutic Application of Simvastatin for Brain Complications and Mechanisms of Action. Pharmaceuticals (Basel) 2023; 16:914. [PMID: 37513826 PMCID: PMC10385015 DOI: 10.3390/ph16070914] [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: 05/08/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 07/30/2023] Open
Abstract
Statins are common drugs that are clinically used to reduce elevated plasma cholesterol levels. Based on their solubility, statins are considered to be either hydrophilic or lipophilic. Amongst them, simvastatin has the highest lipophilicity to facilitate its ability to cross the blood-brain barrier. Recent studies have suggested that simvastatin could be a promising therapeutic option for different brain complications and diseases ranging from brain tumors (i.e., medulloblastoma and glioblastoma) to neurological disorders (i.e., Alzheimer's disease, Parkinson's disease, and Huntington's disease). Specific mechanisms of disease amelioration, however, are still unclear. Independent studies suggest that simvastatin may reduce the risk of developing certain neurodegenerative disorders. Meanwhile, other studies point towards inducing cell death in brain tumor cell lines. In this review, we outline the potential therapeutic effects of simvastatin on brain complications and review the clinically relevant molecular mechanisms in different cases.
Collapse
Affiliation(s)
| | | | - Mojgan Rastegar
- Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
6
|
Rajasekaran S, Ramaian Santhaseela A, Ragunathan S, Venkataraman S, Jayavelu T. Altered Lysosomal Function Manipulates Cellular Biosynthetic Capacity By Remodeling Intracellular Cholesterol Distribution. Cell Biochem Biophys 2023; 81:29-38. [PMID: 36459362 DOI: 10.1007/s12013-022-01123-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022]
Abstract
Lysosomes are known to influence cholesterol trafficking into endoplasmic reticulum (ER) membranes. Though intracellular cholesterol levels are known to influence the lipid biosynthetic responses in ER, the specific effects of lysosomal modulation on these outcomes is not known. To demonstrate this, C2C12 cells were treated with chloroquine, a lysosomotropic agent, and its effects on cellular biosynthetic capacity, structural and functional status of ER was determined. In addition to its known effects on autophagy reduction, chloroquine treatment induced accumulation of total cellular lipid and ER-specific cholesterol content. It was also observed that chloroquine caused an increase in smooth-ER content with defects in overall protein turnover. Further, since ER and mitochondria function in close association through ER membrane contact sites, it is likely that lysosomal modulation also brings about associated changes in mitochondria. In this regard, we found that chloroquine reduces mitochondrial membrane potential and mitochondrial dynamics. Collectively, the differential biosynthetic response of rise in lipid content, but not protein content, cannot be accounted by merely considering that chloroquine induced suppression of autophagy causes defects in organelle function. In this defective autophagy scenario, both biosynthetic responses such as lipid and protein synthesis are expected to be reduced rather than only the latter, as observed with chloroquine. These findings suggest that cholesterol trafficking/distribution within cellular organelles could act as an intracellular mediator of differential biosynthetic remodelling in interconnected organelles.
Collapse
|
7
|
Päth G, Perakakis N, Mantzoros CS, Seufert J. PCSK9 inhibition and cholesterol homeostasis in insulin producing β-cells. Lipids Health Dis 2022; 21:138. [PMID: 36527064 PMCID: PMC9756761 DOI: 10.1186/s12944-022-01751-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Low-density lipoprotein cholesterol (LDL-C) plays a central role in the pathology of atherosclerotic cardiovascular disease. For decades, the gold standard for LDL-C lowering have been statins, although these drugs carry a moderate risk for the development of new-onset diabetes. The inhibitors of proprotein convertase subtilisin/kexin type 9 (PCSK9) have emerged in the last years as potential alternatives to statins due to their high efficiency and safety without indications for a diabetes risk so far. Both approaches finally eliminate LDL-C from bloodstream by upregulation of LDL receptor surface expression. Due to their low antioxidant capacity, insulin producing pancreatic β-cells are sensitive to increased lipid oxidation and related generation of reactive oxygen species. Thus, PCSK9 inhibition has been argued to promote diabetes like statins. Potentially, the remaining patients at risk will be identified in the future. Otherwise, there is increasing evidence that loss of circulating PCSK9 does not worsen glycaemia since it is compensated by local PCSK9 expression in β-cells and other islet cells. This review explores the situation in β-cells. We evaluated the relevant biology of PCSK9 and the effects of its functional loss in rodent knockout models, carriers of LDL-lowering gene variants and PCSK9 inhibitor-treated patients.
Collapse
Affiliation(s)
- Günter Päth
- grid.5963.9Division of Endocrinology and Diabetology, Department of Medicine II, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Germany, Hugstetter Str. 55, Freiburg, Germany
| | - Nikolaos Perakakis
- grid.4488.00000 0001 2111 7257Division of Metabolic and Vascular Medicine, Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany ,grid.38142.3c000000041936754XDivision of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA USA
| | - Christos S. Mantzoros
- grid.38142.3c000000041936754XDivision of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA USA ,grid.410370.10000 0004 4657 1992Section of Endocrinology, VA Boston Healthcare System, MA Jamaica Plain, USA
| | - Jochen Seufert
- grid.5963.9Division of Endocrinology and Diabetology, Department of Medicine II, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Germany, Hugstetter Str. 55, Freiburg, Germany
| |
Collapse
|
8
|
Araki M, Nakagawa Y, Saito H, Yamada Y, Han SI, Mizunoe Y, Ohno H, Miyamoto T, Sekiya M, Matsuzaka T, Sone H, Shimano H. Hepatocyte- or macrophage-specific SREBP-1a deficiency in mice exacerbates methionine- and choline-deficient diet-induced nonalcoholic fatty liver disease. Am J Physiol Gastrointest Liver Physiol 2022; 323:G627-G639. [PMID: 36283088 DOI: 10.1152/ajpgi.00090.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Sterol regulatory element-binding proteins (SREBPs) are master transcription factors for lipid synthesis, and SREBP-1 is important for fatty acid and triglyceride synthesis. SREBP-1 has two isoforms, SREBP-1a and SREBP-1c, which are splicing variants transcribed from the Srebf1 gene. Although SREBP-1a exhibits stronger transcriptional activity than SREBP-1c, hepatic SREBP-1c is considered more physiologically important. We generated SREBP-1a flox mice using the CRISPR/Cas9 system and hepatocyte- and macrophage-specific SREBP-1a knockout (KO) mice (LKO, liver-knockout; and mΦKO, macrophage-knockout). There were no significant differences among all the mouse genotypes upon feeding with a normal diet. However, feeding with a methionine- and choline-deficient (MCD) diet resulted in exacerbated liver injury in both KO mice. In LKO mice, fatty liver was unexpectedly exacerbated, leading to macrophage infiltration and inflammation. In contrast, in mΦKO mice, the fatty liver state was similar to that in flox mice, but the polarity of the macrophages in the liver was transformed into a proinflammatory M1 subtype, resulting in the exacerbation of inflammation. Taken together, we found that SREBP-1a does not contribute to hepatic lipogenesis, but in either hepatocytes or macrophages distinctly controls the onset of pathological conditions in MCD diet-induced hepatitis.NEW & NOTEWORTHY Hepatocyte- and macrophage-specific SREBP-1a knockout mice were generated for the first time. This study reveals that SREBP-1a does not contribute to hepatic lipogenesis, but in either hepatocytes or macrophages distinctly controls the onset of pathological conditions in methionine- and choline-deficient diet-induced hepatitis.
Collapse
Affiliation(s)
- Masaya Araki
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yoshimi Nakagawa
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Division of Complex Biosystem Research, Department of Research and Development, Institute of Natural Medicine, University of Toyama, Toyama, Japan.,Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
| | - Hodaka Saito
- Division of Complex Biosystem Research, Department of Research and Development, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Yasunari Yamada
- Division of Complex Biosystem Research, Department of Research and Development, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Song-Iee Han
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Yuhei Mizunoe
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hiroshi Ohno
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Takafumi Miyamoto
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Motohiro Sekiya
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Takashi Matsuzaka
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Transborder Medical Research Center (TMRC), University of Tsukuba, Tsukuba, Japan
| | - Hirohito Sone
- Department of Hematology, Endocrinology and Metabolism, Faculty of Medicine, Niigata University, Niigata, Japan
| | - Hitoshi Shimano
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan.,International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan.,Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| |
Collapse
|
9
|
Dai J, Wang H, Liao Y, Tan L, Sun Y, Song C, Liu W, Qiu X, Ding C. Coronavirus Infection and Cholesterol Metabolism. Front Immunol 2022; 13:791267. [PMID: 35529872 PMCID: PMC9069556 DOI: 10.3389/fimmu.2022.791267] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 03/21/2022] [Indexed: 12/19/2022] Open
Abstract
Host cholesterol metabolism remodeling is significantly associated with the spread of human pathogenic coronaviruses, suggesting virus-host relationships could be affected by cholesterol-modifying drugs. Cholesterol has an important role in coronavirus entry, membrane fusion, and pathological syncytia formation, therefore cholesterol metabolic mechanisms may be promising drug targets for coronavirus infections. Moreover, cholesterol and its metabolizing enzymes or corresponding natural products exert antiviral effects which are closely associated with individual viral steps during coronavirus replication. Furthermore, the coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 infections are associated with clinically significant low cholesterol levels, suggesting cholesterol could function as a potential marker for monitoring viral infection status. Therefore, weaponizing cholesterol dysregulation against viral infection could be an effective antiviral strategy. In this review, we comprehensively review the literature to clarify how coronaviruses exploit host cholesterol metabolism to accommodate viral replication requirements and interfere with host immune responses. We also focus on targeting cholesterol homeostasis to interfere with critical steps during coronavirus infection.
Collapse
Affiliation(s)
- Jun Dai
- College of Animal Science and Technology, Guangxi University, Nanning, China
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Experimental Animal Center, Zunyi Medical University, Zunyi City, China
| | - Huan Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Ying Liao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Lei Tan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Yingjie Sun
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Cuiping Song
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Weiwei Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Xusheng Qiu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- *Correspondence: Xusheng Qiu, ; Chan Ding,
| | - Chan Ding
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- *Correspondence: Xusheng Qiu, ; Chan Ding,
| |
Collapse
|
10
|
Luquero A, Vilahur G, Casani L, Badimon L, Borrell-Pages M. Differential cholesterol uptake in liver cells: A role for PCSK9. FASEB J 2022; 36:e22291. [PMID: 35344222 DOI: 10.1096/fj.202101660rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/11/2022] [Accepted: 03/20/2022] [Indexed: 12/21/2022]
Abstract
The clearance of low-density lipoprotein (LDL) particles from the circulation is regulated by the LDL receptor (LDLR) and proprotein convertase subtilisin/kexin 9 (PCSK9) interaction. Its disruption reduces blood cholesterol levels and delays atherosclerosis progression. Whether other members of the LDLR superfamily are in vivo targets of PCSK9 has been poorly explored. The aim of this work was to study the interaction between PCSK9 and members of the LDLR superfamily in the regulation of liver cholesterol homeostasis in an in vivo low-density lipoprotein receptor related protein 5 (LRP5) deficient mice model challenged with high-fat diet. Our results show that Wt and Lrp5-/- mice fed a hypercholesterolemic diet (HC) have increased cholesterol ester accumulation and decreased liver LDLR and LRP5 gene and protein expression. Very low-density lipoprotein receptor (VLDLR), LRP6, LRP2, and LRP1 expression levels were analyzed in liver samples and show that they do not participate in Lrp5-/- liver cholesterol uptake. Immunoprecipitation experiments show that LRP5 forms a complex with PCSK9 in liver-specific fat-storing stellate cells but not in structural HepG2 cells. Hepatic stellate cells silenced for LRP5 and/or PCSK9 expression and challenged with lipids show reduced cholesterol ester accumulation, indicating that both proteins are involved in lipid processing in the liver. Our results indicate that cholesterol esters accumulate in livers of Wt mice in a LDLR-family-members dependent manner as VLDLR, LRP2, and LRP6 show increased expression in HC mice. However, this increase is lost in livers of Lrp5-/- mice, where scavenger receptors are involved in cholesterol uptake. PCSK9 expression is strongly downregulated in mice livers after HC feeding. However PCSK9 and LRP5 bind in the cytoplasm of fat storing liver cells, indicating that this PCSK9-LRP5 interaction is cell-type specific and that both proteins contribute to lipid uptake.
Collapse
Affiliation(s)
- Aureli Luquero
- Cardiovascular Program ICCC, Hospital de la Santa Creu i Sant Pau Research Institute, IIB-Sant Pau, Barcelona, Spain
| | - Gemma Vilahur
- Cardiovascular Program ICCC, Hospital de la Santa Creu i Sant Pau Research Institute, IIB-Sant Pau, Barcelona, Spain.,CIBER-CV, Instituto de Salud Carlos III, Spain
| | - Laura Casani
- Cardiovascular Program ICCC, Hospital de la Santa Creu i Sant Pau Research Institute, IIB-Sant Pau, Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Program ICCC, Hospital de la Santa Creu i Sant Pau Research Institute, IIB-Sant Pau, Barcelona, Spain.,CIBER-CV, Instituto de Salud Carlos III, Spain.,Cardiovascular Research Chair, UAB, Barcelona, Spain
| | - Maria Borrell-Pages
- Cardiovascular Program ICCC, Hospital de la Santa Creu i Sant Pau Research Institute, IIB-Sant Pau, Barcelona, Spain.,CIBER-CV, Instituto de Salud Carlos III, Spain
| |
Collapse
|
11
|
Takeuchi Y, Yahagi N, Aita Y, Mehrazad-Saber Z, Ho MH, Huyan Y, Murayama Y, Shikama A, Masuda Y, Izumida Y, Miyamoto T, Matsuzaka T, Kawakami Y, Shimano H. FoxO-KLF15 pathway switches the flow of macronutrients under the control of insulin. iScience 2021; 24:103446. [PMID: 34988390 PMCID: PMC8710527 DOI: 10.1016/j.isci.2021.103446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 09/18/2021] [Accepted: 11/11/2021] [Indexed: 11/15/2022] Open
Abstract
KLF15 is a transcription factor that plays an important role in the activation of gluconeogenesis from amino acids as well as the suppression of lipogenesis from glucose. Here we identified the transcription start site of liver-specific KLF15 transcript and showed that FoxO1/3 transcriptionally regulates Klf15 gene expression by directly binding to the liver-specific Klf15 promoter. To achieve this, we performed a precise in vivo promoter analysis combined with the genome-wide transcription-factor-screening method "TFEL scan", using our original Transcription Factor Expression Library (TFEL), which covers nearly all the transcription factors in the mouse genome. Hepatic Klf15 expression is significantly increased via FoxOs by attenuating insulin signaling. Furthermore, FoxOs elevate the expression levels of amino acid catabolic enzymes and suppress SREBP-1c via KLF15, resulting in accelerated amino acid breakdown and suppressed lipogenesis during fasting. Thus, the FoxO-KLF15 pathway contributes to switching the macronutrient flow in the liver under the control of insulin.
Collapse
Affiliation(s)
- Yoshinori Takeuchi
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Naoya Yahagi
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yuichi Aita
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Zahra Mehrazad-Saber
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Man Hei Ho
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yiren Huyan
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Yuki Murayama
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Akito Shikama
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yukari Masuda
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yoshihiko Izumida
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Takafumi Miyamoto
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Takashi Matsuzaka
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yasushi Kawakami
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Hitoshi Shimano
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
12
|
Park G, Jung S, Wellen KE, Jang C. The interaction between the gut microbiota and dietary carbohydrates in nonalcoholic fatty liver disease. Exp Mol Med 2021; 53:809-822. [PMID: 34017059 PMCID: PMC8178320 DOI: 10.1038/s12276-021-00614-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/24/2021] [Indexed: 02/04/2023] Open
Abstract
Imbalance between fat production and consumption causes various metabolic disorders. Nonalcoholic fatty liver disease (NAFLD), one such pathology, is characterized by abnormally increased fat synthesis and subsequent fat accumulation in hepatocytes1,2. While often comorbid with obesity and insulin resistance, this disease can also be found in lean individuals, suggesting specific metabolic dysfunction2. NAFLD has become one of the most prevalent liver diseases in adults worldwide, but its incidence in both children and adolescents has also markedly increased in developed nations3,4. Progression of this disease into nonalcoholic steatohepatitis (NASH), cirrhosis, liver failure, and hepatocellular carcinoma in combination with its widespread incidence thus makes NAFLD and its related pathologies a significant public health concern. Here, we review our understanding of the roles of dietary carbohydrates (glucose, fructose, and fibers) and the gut microbiota, which provides essential carbon sources for hepatic fat synthesis during the development of NAFLD.
Collapse
Affiliation(s)
- Grace Park
- Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | - Sunhee Jung
- Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
13
|
Funes AK, Simón L, Colombo R, Avena MV, Monclús M, Crescitelli J, Cabrillana ME, Conte MI, Cayado N, Boarelli P, Fornés MW, Saez Lancellotti TE. Impact of high fat diet on the sterol regulatory element-binding protein 2 cholesterol pathway in the testicle. Mol Hum Reprod 2021; 27:6206393. [PMID: 33787903 DOI: 10.1093/molehr/gaab023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 03/18/2021] [Indexed: 12/30/2022] Open
Abstract
Male fertility has been shown to be dependent on cholesterol homeostasis. This lipid is essential for testosterone synthesis and spermatogenesis, but its levels must be maintained in an optimal range for proper testicular function. In particular, sperm cells' development is very sensitive to high cholesterol levels, noticeably during acrosomal formation. The aim of this work was to study whether the molecular pathway that regulates intracellular cholesterol, the sterol regulatory element-binding protein (SREBP) pathway, is affected in the testicles of animals under a fat diet. To investigate this, we took advantage of the non-obese hypercholesterolemia (HC) model in New Zealand rabbits that displays poor sperm and seminal quality. The testicular expression of SREBP isoform 2 (SREBP2) and its target molecules 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGCR) and low-density lipoprotein receptor (LDLR) were studied under acute (6 months) and chronic (more than 12 months) fat intake by RT-PCR, western blot and immunofluorescence. Our findings showed that fat consumption promoted down-regulation of the SREBP2 pathway in the testicle at 6 months, but upregulation after a chronic period. This was consistent with load of testicular cholesterol, assessed by filipin staining. In conclusion, the intracellular pathway that regulates cholesterol levels in the testicle is sensitive to dietary fats, and behaves differently depending on the duration of consumption: it has a short-term protective effect, but became deregulated in the long term, ultimately leading to a detrimental situation. These results will contribute to the understanding of the basic mechanisms of the effect of fat consumption in humans with idiopathic infertility.
Collapse
Affiliation(s)
- Abi K Funes
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Laboratorio de Biología Molecular del Metabolismo & Nutrición (bMeNu)†, Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - Layla Simón
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - Regina Colombo
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Laboratorio de Biología Molecular del Metabolismo & Nutrición (bMeNu)†, Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - María Virginia Avena
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Laboratorio de Biología Molecular del Metabolismo & Nutrición (bMeNu)†, Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - María Monclús
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Instituto de Investigaciones. Facultad de Ciencias Médicas. Universidad del Aconcagua. Mendoza, Argentina
| | - Julieta Crescitelli
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (bMeNu)†, Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Instituto de Investigaciones. Facultad de Ciencias Médicas. Universidad del Aconcagua. Mendoza, Argentina
| | - María E Cabrillana
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - María Inés Conte
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - Niubys Cayado
- Instituto de Investigaciones. Facultad de Ciencias Médicas. Universidad del Aconcagua. Mendoza, Argentina.,Laboratorio de Oncología, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - Paola Boarelli
- Laboratorio de Enfermedades Metabólicas (LEM), Universidad Juan Agustín Maza, Mendoza, Argentina
| | - Miguel W Fornés
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - Tania E Saez Lancellotti
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Laboratorio de Biología Molecular del Metabolismo & Nutrición (bMeNu)†, Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Instituto de Investigaciones. Facultad de Ciencias Médicas. Universidad del Aconcagua. Mendoza, Argentina
| |
Collapse
|
14
|
High dose rosuvastatin increases ABCA1 transporter in human atherosclerotic plaques in a cholesterol-independent fashion. Int J Cardiol 2019; 299:249-253. [PMID: 31409515 DOI: 10.1016/j.ijcard.2019.07.094] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 07/10/2019] [Accepted: 07/30/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND ATP-binding cassette A1 (ABCA1) and G1 (ABCG1) mediate cholesterol efflux from lipid-laden macrophages, thus promoting anti-atherosclerotic outcomes. The mechanism(s) linking treatment with statins and ABCA1/ABCG1 in human atherosclerosis are not fully understood and require further investigation. Therefore, we studied whether short-term treatment with low- or high-dose rosuvastatin may affect ABCA1 and ABCG1 expression in human atherosclerotic plaques. METHODS Seventy patients with severe stenosis of the internal carotid artery were randomized to receive low (10 mg/day) or high (40 mg/day) dose rosuvastatin for 12 weeks before elective endarterectomy. As controls, we analyzed a reference group of 10 plaques from subjects with hypercholesterolemia but not receiving statin treatment and an additional set of 11 plaques collected from normocholesterolemic patients. On atherosclerotic plaques, ABCA1 and ABCG1 expression was evaluated at RNA level by qPCR and at protein level by immunoblotting and immunohistochemistry. RESULTS Both rosuvastatin doses were associated with lower plaque ABCA1 mRNA levels and with a trend toward reduction for ABCG1. However, ABCA1 protein was paradoxically higher in patients treated with high-dose rosuvastatin and was associated with lower levels of miR-33b-5p, a microRNA known as a regulator of ABCA1. Multivariate analyses showed that the effect is cholesterol-independent. Finally, no effects were found for ABCG1 protein. CONCLUSIONS High-dose rosuvastatin increases macrophage ABCA1 protein levels in human atherosclerotic plaque despite mRNA reduction in a mechanism unrelated to plasma cholesterol reduction and potentially involving miR-33b-5p. This pathway may reflect an additional feature contributing to the anti-atherosclerotic effect for high-dose rosuvastatin. TRIAL REGISTRATION ISRCTN16590640.
Collapse
|
15
|
Galvan AM, Chorba JS. Cell-associated heparin-like molecules modulate the ability of LDL to regulate PCSK9 uptake. J Lipid Res 2018; 60:71-84. [PMID: 30463987 DOI: 10.1194/jlr.m087189] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 10/30/2018] [Indexed: 01/12/2023] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) targets the LDL receptor (LDLR) for degradation, increasing plasma LDL and, consequently, cardiovascular risk. Uptake of secreted PCSK9 is required for its effect on the LDLR, and LDL itself inhibits this uptake, though how it does so remains unclear. In this study, we investigated the relationship between LDL, the PCSK9:LDLR interaction, and PCSK9 uptake. We show that LDL inhibits binding of PCSK9 to the LDLR in vitro more impressively than it inhibits PCSK9 uptake in cells. Furthermore, cell-surface heparin-like molecules (HLMs) can partly explain this difference, consistent with heparan sulfate proteoglycans (HSPGs) acting as coreceptors for PCSK9. We also show that HLMs can interact with either PCSK9 or LDL to modulate the inhibitory activity of LDL on PCSK9 uptake, with such inhibition rescued by competition with the entire PCSK9 prodomain, but not its truncated variants. Additionally, we show that the gain-of-function PCSK9 variant, S127R, located in the prodomain near the HSPG binding site, exhibits increased affinity for HLMs, potentially explaining its phenotype. Overall, our findings suggest a model where LDL acts as a negative regulator of PCSK9 function by decreasing its uptake via direct interactions with either the LDLR or HLMs.
Collapse
Affiliation(s)
- Adri M Galvan
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California San Francisco, San Francisco, CA 94143
| | - John S Chorba
- Division of Cardiology, Zuckerberg San Francisco General, Department of Medicine, University of California San Francisco, San Francisco, CA 94110
| |
Collapse
|
16
|
Simón L, Funes AK, Monclús MA, Colombo R, Cabrillana ME, Saez Lancellotti TE, Fornés MW. Manchette-acrosome disorders and testicular efficiency decline observed in hypercholesterolemic rabbits are recovered with olive oil enriched diet. PLoS One 2018; 13:e0202748. [PMID: 30138421 PMCID: PMC6107225 DOI: 10.1371/journal.pone.0202748] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 08/08/2018] [Indexed: 12/04/2022] Open
Abstract
High-fat diet is associated with hypercholesterolemia and seminal alterations in White New Zealand rabbits. We have previously reported disorders in the development of the manchette-acrosome complex during spermiogenesis and decreased testicular efficiency in hypercholesterolemic rabbits. On the other hand, olive oil incorporated into the diet improves cholesterolemia and semen parameters affected in hypercholesterolemic rabbits. In this paper, we report the recovery—with the addition of olive oil to diet—from the sub-cellular mechanisms involved in the shaping of the sperm cell and testicular efficiency altered in hypercholesterolemic rabbits. Using morphological (structural, ultra-structural and immuno-fluorescence techniques) and cell biology techniques, a reorganization of the manchette and related structures was observed when olive oil was added to the high-fat diet. Specifically, actin filaments, microtubules and lipid rafts—abnormally distributed in hypercholesterolemic rabbits—were recovered with dietary olive oil supplementation. The causes of the decline in sperm count were studied in the previous report and here in more detail. These were attributed to the decrease in the efficiency index and also to the increase in the apoptotic percentage in testis from animals under the high-fat diet. Surprisingly, the addition of olive oil to the diet avoided the sub-cellular, efficiency and apoptosis changes observed in hypercholesterolemic rabbits. This paper reports the positive effects of the olive oil addition to the diet in the recovery of testicular efficiency and normal sperm shaping, mechanisms altered by hypercholesterolemia.
Collapse
Affiliation(s)
- Layla Simón
- IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- * E-mail:
| | - Abi K. Funes
- IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
| | - María A. Monclús
- IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Instituto de investigaciones, Facultad de Ciencias Médicas, Universidad del Aconcagua, Mendoza, Argentina
| | - Regina Colombo
- IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
| | - María E. Cabrillana
- IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Instituto de investigaciones, Facultad de Ciencias Médicas, Universidad del Aconcagua, Mendoza, Argentina
| | - Tania E. Saez Lancellotti
- IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Instituto de investigaciones, Facultad de Ciencias Médicas, Universidad del Aconcagua, Mendoza, Argentina
| | - Miguel W. Fornés
- IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Instituto de investigaciones, Facultad de Ciencias Médicas, Universidad del Aconcagua, Mendoza, Argentina
| |
Collapse
|
17
|
Lei MM, Wei CK, Chen Z, Yosefi S, Zhu HX, Shi ZD. Anti-leptin receptor antibodies strengthen leptin biofunction in growing chickens. Gen Comp Endocrinol 2018; 259:223-230. [PMID: 29247679 DOI: 10.1016/j.ygcen.2017.12.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 12/13/2017] [Accepted: 12/13/2017] [Indexed: 12/11/2022]
Abstract
Antibodies against the extracellular domains of the chicken leptin receptor were used to study the biological function of leptin in growing chickens. Both polyclonal and monoclonal anti-LEPR antibodies were administered intramuscularly to 30-d-old Chinese indigenous Gushi pullets. Both antibody preparations increased feed intake for 6 h after injection and reduced plasma concentrations of glucose, triglycerides, and both high- and low-density lipoproteins. The antibody treatments also upregulated agouti-related peptide and neuropeptide Y in the hypothalamus and downregulated proopiomelanocortin, melanocortin 4 receptor, and leptin receptor. The treatments also upregulated leptin receptor, acetyl CoA carboxylase beta, and acyl-CoA oxidase in the liver, abdominal fat, and breast muscle and downregulated sterol regulatory element-binding protein-1 and fatty acid synthase. Furthermore, even though the anti-leptin receptor antibodies failed to affect leptin receptor signaling transduction when administered alone, they did augment the induction of leptin receptor signaling transduction by leptin. These results demonstrate that antibodies against the extracellular domains of leptin-specific receptor enhance, but do not mimic, the ability of leptin to activate receptors. Furthermore, the enhanced leptin bioactivity observed after the intramuscular injection of anti-LEPR antibodies confirmed the occurrence of de novo leptin in the peripheral tissues and blood of treated chickens.
Collapse
Affiliation(s)
- M M Lei
- Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality, MOA, Nanjing 210014, China; Laboratory of Animal Breeding and Reproduction, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.
| | - C K Wei
- Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality, MOA, Nanjing 210014, China; Laboratory of Animal Breeding and Reproduction, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.
| | - Z Chen
- Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality, MOA, Nanjing 210014, China; Laboratory of Animal Breeding and Reproduction, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.
| | - S Yosefi
- Institute of Animal Science, Agricultural Research Organization, Volcani Center, PO Box 6, Bet Dagan 50250, Israel.
| | - H X Zhu
- Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality, MOA, Nanjing 210014, China; Laboratory of Animal Breeding and Reproduction, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.
| | - Z D Shi
- Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality, MOA, Nanjing 210014, China; Laboratory of Animal Breeding and Reproduction, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.
| |
Collapse
|
18
|
Rong X, Wang B, Palladino EN, de Aguiar Vallim TQ, Ford DA, Tontonoz P. ER phospholipid composition modulates lipogenesis during feeding and in obesity. J Clin Invest 2017; 127:3640-3651. [PMID: 28846071 DOI: 10.1172/jci93616] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 07/11/2017] [Indexed: 12/31/2022] Open
Abstract
Sterol regulatory element-binding protein 1c (SREBP-1c) is a central regulator of lipogenesis whose activity is controlled by proteolytic cleavage. The metabolic factors that affect its processing are incompletely understood. Here, we show that dynamic changes in the acyl chain composition of ER phospholipids affect SREBP-1c maturation in physiology and disease. The abundance of polyunsaturated phosphatidylcholine in liver ER is selectively increased in response to feeding and in the setting of obesity-linked insulin resistance. Exogenous delivery of polyunsaturated phosphatidylcholine to ER accelerated SREBP-1c processing through a mechanism that required an intact SREBP cleavage-activating protein (SCAP) pathway. Furthermore, induction of the phospholipid-remodeling enzyme LPCAT3 in response to liver X receptor (LXR) activation promoted SREBP-1c processing by driving the incorporation of polyunsaturated fatty acids into ER. Conversely, LPCAT3 deficiency increased membrane saturation, reduced nuclear SREBP-1c abundance, and blunted the lipogenic response to feeding, LXR agonist treatment, or obesity-linked insulin resistance. Desaturation of the ER membrane may serve as an auxiliary signal of the fed state that promotes lipid synthesis in response to nutrient availability.
Collapse
Affiliation(s)
- Xin Rong
- Department of Pathology and Laboratory Medicine, Department of Medicine, UCLA, Los Angeles, California, USA
| | - Bo Wang
- Department of Pathology and Laboratory Medicine, Department of Medicine, UCLA, Los Angeles, California, USA
| | - Elisa Nd Palladino
- Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University, St. Louis, Missouri, USA
| | | | - David A Ford
- Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University, St. Louis, Missouri, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, Department of Medicine, UCLA, Los Angeles, California, USA.,Molecular Biology Institute, and.,Howard Hughes Medical Institute, UCLA, Los Angeles, California, USA
| |
Collapse
|
19
|
Silvagno F, Pescarmona G. Spotlight on vitamin D receptor, lipid metabolism and mitochondria: Some preliminary emerging issues. Mol Cell Endocrinol 2017; 450:24-31. [PMID: 28414049 DOI: 10.1016/j.mce.2017.04.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 04/07/2017] [Accepted: 04/09/2017] [Indexed: 12/19/2022]
Abstract
Transcriptional control and modulation of calcium fluxes underpin the differentiating properties of vitamin D (1,25(OH)2D3). In the latest years however few studies have pointed out the relevance of the mitochondrial effects of the hormone. It is now time to focus on the metabolic results of vitamin D receptor (VDR) action in mitochondria, which can explain the pleiotropic effects of 1,25(OH)2D3 and may elucidate few contrasting aspects of its activity. The perturbation of lipid metabolism described in VDR knockout mice and vitamin D deficient animals can be revisited based on the newly identified mechanism of action of 1,25(OH)2D3 in mitochondria. From the same point of view, the controversial role of 1,25(OH)2D3 in adipogenesis can be better interpreted.
Collapse
Affiliation(s)
- Francesca Silvagno
- Department of Oncology, University of Torino, Via Santena 5 bis, 10126 Torino, Italy.
| | - Gianpiero Pescarmona
- Department of Oncology, University of Torino, Via Santena 5 bis, 10126 Torino, Italy.
| |
Collapse
|
20
|
Udata C, Garzone PD, Gumbiner B, Joh T, Liang H, Liao KH, Williams JH, Meng X. A Mechanism-Based Pharmacokinetic/Pharmacodynamic Model for Bococizumab, a Humanized Monoclonal Antibody Against Proprotein Convertase Subtilisin/Kexin Type 9, and Its Application in Early Clinical Development. J Clin Pharmacol 2017; 57:855-864. [DOI: 10.1002/jcph.867] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 12/05/2016] [Indexed: 12/15/2022]
Affiliation(s)
| | - Pamela D. Garzone
- World Wide Research and Development; Pfizer Inc; San Diego California
| | - Barry Gumbiner
- World Wide Research and Development; Pfizer Inc; San Diego California
| | - Tenshang Joh
- World Wide Research and Development; Pfizer Inc; San Diego California
| | - Hong Liang
- World Wide Research and Development; Pfizer Inc; San Diego California
| | - Kai-Hsin Liao
- World Wide Research and Development; Pfizer Inc; San Diego California
| | - Jason H. Williams
- World Wide Research and Development; Pfizer Inc; San Diego California
| | - Xu Meng
- World Wide Research and Development; Pfizer Inc; San Diego California
| |
Collapse
|
21
|
Takeuchi Y, Yahagi N, Aita Y, Murayama Y, Sawada Y, Piao X, Toya N, Oya Y, Shikama A, Takarada A, Masuda Y, Nishi M, Kubota M, Izumida Y, Yamamoto T, Sekiya M, Matsuzaka T, Nakagawa Y, Urayama O, Kawakami Y, Iizuka Y, Gotoda T, Itaka K, Kataoka K, Nagai R, Kadowaki T, Yamada N, Lu Y, Jain MK, Shimano H. KLF15 Enables Rapid Switching between Lipogenesis and Gluconeogenesis during Fasting. Cell Rep 2016; 16:2373-86. [PMID: 27545894 PMCID: PMC5031553 DOI: 10.1016/j.celrep.2016.07.069] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 07/08/2016] [Accepted: 07/25/2016] [Indexed: 11/17/2022] Open
Abstract
Hepatic lipogenesis is nutritionally regulated (i.e., downregulated during fasting and upregulated during the postprandial state) as an adaptation to the nutritional environment. While alterations in the expression level of the transcription factor SREBP-1c are known to be critical for nutritionally regulated lipogenesis, upstream mechanisms governing Srebf1 expression remain unclear. Here, we show that the fasting-induced transcription factor KLF15, a key regulator of gluconeogenesis, forms a complex with LXR/RXR, specifically on the Srebf1 promoter. This complex recruits the corepressor RIP140 instead of the coactivator SRC1, resulting in reduced Srebf1 and thus downstream lipogenic enzyme expression during the early and euglycemic period of fasting prior to hypoglycemia and PKA activation. Through this mechanism, KLF15 overexpression specifically ameliorates hypertriglyceridemia without affecting LXR-mediated cholesterol metabolism. These findings reveal a key molecular link between glucose and lipid metabolism and have therapeutic implications for the treatment of hyperlipidemia.
Collapse
Affiliation(s)
- Yoshinori Takeuchi
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Naoya Yahagi
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan.
| | - Yuichi Aita
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Yuki Murayama
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Yoshikazu Sawada
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Xiaoying Piao
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Naoki Toya
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Yukari Oya
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Akito Shikama
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Ayako Takarada
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Yukari Masuda
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Makiko Nishi
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Midori Kubota
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Yoshihiko Izumida
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Takashi Yamamoto
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Motohiro Sekiya
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Takashi Matsuzaka
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Yoshimi Nakagawa
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Osamu Urayama
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Yasushi Kawakami
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Yoko Iizuka
- Department of Internal Medicine, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Takanari Gotoda
- Department of Internal Medicine, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Keiji Itaka
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Kazunori Kataoka
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Ryozo Nagai
- Department of Internal Medicine, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Takashi Kadowaki
- Department of Internal Medicine, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Nobuhiro Yamada
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Yuan Lu
- Case Cardiovascular Research Institute, Cleveland, OH 44106, USA
| | - Mukesh K Jain
- Case Cardiovascular Research Institute, Cleveland, OH 44106, USA
| | - Hitoshi Shimano
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
22
|
Karahashi M, Hirata-Hanta Y, Kawabata K, Tsutsumi D, Kametani M, Takamatsu N, Sakamoto T, Yamazaki T, Asano S, Mitsumoto A, Kawashima Y, Kudo N. Abnormalities in the Metabolism of Fatty Acids and Triacylglycerols in the Liver of the Goto-Kakizaki Rat: A Model for Non-Obese Type 2 Diabetes. Lipids 2016; 51:955-71. [PMID: 27372943 DOI: 10.1007/s11745-016-4171-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 06/20/2016] [Indexed: 02/08/2023]
Abstract
The Goto-Kakizaki (GK) rat is widely used as an animal model for spontaneous-onset type 2 diabetes without obesity; nevertheless, little information is available on the metabolism of fatty acids and triacylglycerols (TAG) in their livers. We investigated the mechanisms underlying the alterations in the metabolism of fatty acids and TAG in their livers, in comparison with Zucker (fa/fa) rats, which are obese and insulin resistant. Lipid profiles, the expression of genes for enzymes and proteins related to the metabolism of fatty acid and TAG, de novo synthesis of fatty acids and TAG in vivo, fatty acid synthase activity in vitro, fatty acid oxidation in liver slices, and very-low-density-lipoprotein (VLDL)-TAG secretion in vivo were estimated. Our results revealed that (1) the TAG accumulation was moderate, (2) the de novo fatty acid synthesis was increased by upregulation of fatty acid synthase in a post-transcriptional manner, (3) fatty acid oxidation was also augmented through the induction of carnitine palmitoyltransferase 1a, and (4) the secretion rate of VLDL-TAG remained unchanged in the livers of GK rats. These results suggest that, despite the fact that GK rats exhibit non-obese type 2 diabetes, the upregulation of de novo lipogenesis is largely compensated by the upregulation of fatty acid oxidation, resulting in only moderate increase in TAG accumulation in the liver.
Collapse
Affiliation(s)
- Minako Karahashi
- School of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Yuko Hirata-Hanta
- School of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Kohei Kawabata
- School of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Daisuke Tsutsumi
- School of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Misaki Kametani
- School of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Nanako Takamatsu
- School of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Takeshi Sakamoto
- School of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Tohru Yamazaki
- School of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Satoshi Asano
- Department of Pharmaceutical Sciences, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi, 324-8501, Japan
| | - Atsushi Mitsumoto
- Faculty of Pharmaceutical Sciences, Josai International University, 1 Gumyo, Togane, Chiba, 283-8555, Japan
| | - Yoichi Kawashima
- School of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Naomi Kudo
- School of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan.
| |
Collapse
|
23
|
Lagace TA. Phosphatidylcholine: Greasing the Cholesterol Transport Machinery. Lipid Insights 2016; 8:65-73. [PMID: 27081313 PMCID: PMC4821435 DOI: 10.4137/lpi.s31746] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 02/26/2016] [Accepted: 02/23/2016] [Indexed: 01/19/2023] Open
Abstract
Negative feedback regulation of cholesterol metabolism in mammalian cells ensures a proper balance of cholesterol with other membrane lipids, principal among these being the major phospholipid phosphatidylcholine (PC). Processes such as cholesterol biosynthesis and efflux, cholesteryl ester storage in lipid droplets, and uptake of plasma lipoproteins are tuned to the cholesterol/PC ratio. Cholesterol-loaded macrophages in atherosclerotic lesions display increased PC biosynthesis that buffers against elevated cholesterol levels and may also facilitate cholesterol trafficking to enhance cholesterol sensing and efflux. These same mechanisms could play a generic role in homeostatic responses to acute changes in membrane free cholesterol levels. Here, I discuss the established and emerging roles of PC metabolism in promoting intracellular cholesterol trafficking and membrane lipid homeostasis.
Collapse
Affiliation(s)
- Thomas A Lagace
- Department of Pathology and Laboratory Medicine, University of Ottawa Heart Institute, Ottawa, ON, Canada
| |
Collapse
|
24
|
McFarlane MR, Cantoria MJ, Linden AG, January BA, Liang G, Engelking LJ. Scap is required for sterol synthesis and crypt growth in intestinal mucosa. J Lipid Res 2015; 56:1560-71. [PMID: 25896350 DOI: 10.1194/jlr.m059709] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Indexed: 11/20/2022] Open
Abstract
SREBP cleavage-activating protein (Scap) is an endoplasmic reticulum membrane protein required for cleavage and activation of sterol regulatory element-binding proteins (SREBPs), which activate the transcription of genes in sterol and fatty acid biosynthesis. Liver-specific loss of Scap is well tolerated; hepatic synthesis of sterols and fatty acids is reduced, but mice are otherwise healthy. To determine whether Scap loss is tolerated in the intestine, we generated a mouse model (Vil-Scap(-)) in which tamoxifen-inducible Cre-ER(T2), a fusion protein of Cre recombinase with a mutated ligand binding domain of the human estrogen receptor, ablates Scap in intestinal mucosa. After 4 days of tamoxifen, Vil-Scap(-) mice succumb with a severe enteropathy and near-complete collapse of intestinal mucosa. Organoids grown ex vivo from intestinal crypts of Vil-Scap(-) mice are readily killed when Scap is deleted by 4-hydroxytamoxifen. Death is prevented when culture medium is supplemented with cholesterol and oleate. These data show that, unlike the liver, the intestine requires Scap to sustain tissue integrity by maintaining the high levels of lipid synthesis necessary for proliferation of intestinal crypts.
Collapse
Affiliation(s)
- Matthew R McFarlane
- Department of Molecular Genetics University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
| | - Mary Jo Cantoria
- Department of Molecular Genetics University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
| | - Albert G Linden
- Department of Molecular Genetics University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
| | - Brandon A January
- Department of Molecular Genetics University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
| | - Guosheng Liang
- Department of Molecular Genetics University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
| | - Luke J Engelking
- Department of Molecular Genetics University of Texas Southwestern Medical Center, Dallas, TX 75390-9046 Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
| |
Collapse
|
25
|
Goldstein JL, Brown MS. A century of cholesterol and coronaries: from plaques to genes to statins. Cell 2015; 161:161-172. [PMID: 25815993 PMCID: PMC4525717 DOI: 10.1016/j.cell.2015.01.036] [Citation(s) in RCA: 793] [Impact Index Per Article: 79.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Indexed: 01/06/2023]
Abstract
One-fourth of all deaths in industrialized countries result from coronary heart disease. A century of research has revealed the essential causative agent: cholesterol-carrying low-density lipoprotein (LDL). LDL is controlled by specific receptors (LDLRs) in liver that remove it from blood. Mutations that eliminate LDLRs raise LDL and cause heart attacks in childhood, whereas mutations that raise LDLRs reduce LDL and diminish heart attacks. If we are to eliminate coronary disease, lowering LDL should be the primary goal. Effective means to achieve this goal are currently available. The key questions are: who to treat, when to treat, and how long to treat.
Collapse
Affiliation(s)
- Joseph L Goldstein
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Michael S Brown
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
26
|
Ono K, Horie T, Nishino T, Baba O, Kuwabara Y, Yokode M, Kita T, Kimura T. MicroRNA-33a/b in lipid metabolism – novel “thrifty” models. Circ J 2015; 79:278-84. [PMID: 25744742 DOI: 10.1253/circj.cj-14-1252] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
MicroRNAs (miRNAs; miRs) are small non-protein-coding RNAs that negatively regulate gene expression. They bind to the 3' UTR of specific mRNAs and either inhibit translation or promote mRNA degradation. There is emerging evidence linking miR-33a/b to lipid homoeostasis, targeting ABCA1,SREBF1, etc and it would appear that they have acted as "thrifty genes" during evolution to maintain cholesterol levels both at the cellular and whole body level. As we are now living in a period of "satiation", miR-33a/b no longer seem to be useful and could be potential therapeutic targets for lipid disorders and/or atherosclerosis. In this review, we describe the current understanding of the function of miR-33a/b in lipid homeostasis, focusing on the "thrifty" aspect.
Collapse
Affiliation(s)
- Koh Ono
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Horie T, Nishino T, Baba O, Kuwabara Y, Nakao T, Nishiga M, Usami S, Izuhara M, Sowa N, Yahagi N, Shimano H, Matsumura S, Inoue K, Marusawa H, Nakamura T, Hasegawa K, Kume N, Yokode M, Kita T, Kimura T, Ono K. MicroRNA-33 regulates sterol regulatory element-binding protein 1 expression in mice. Nat Commun 2014; 4:2883. [PMID: 24300912 PMCID: PMC3863899 DOI: 10.1038/ncomms3883] [Citation(s) in RCA: 175] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Accepted: 11/07/2013] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRs) are small non-protein-coding RNAs that bind to specific mRNAs and inhibit translation or promote mRNA degradation. Recent reports have indicated that miR-33, which is located within the intron of sterol regulatory element-binding protein (SREBP) 2, controls cholesterol homoeostasis and may be a potential therapeutic target for the treatment of atherosclerosis. Here we show that deletion of miR-33 results in marked worsening of high-fat diet-induced obesity and liver steatosis. Using miR-33(-/-)Srebf1(+/-) mice, we demonstrate that SREBP-1 is a target of miR-33 and that the mechanisms leading to obesity and liver steatosis in miR-33(-/-) mice involve enhanced expression of SREBP-1. These results elucidate a novel interaction between SREBP-1 and SREBP-2 mediated by miR-33 in vivo.
Collapse
Affiliation(s)
- Takahiro Horie
- 1] Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan [2] Department of Clinical Innovative Medicine, Institute for Advancement of Clinical and Translational Science, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan [3]
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Altemus JB, Patel SB, Sehayek E. Liver-specific induction of Abcg5 and Abcg8 stimulates reverse cholesterol transport in response to ezetimibe treatment. Metabolism 2014; 63:1334-41. [PMID: 25060694 DOI: 10.1016/j.metabol.2014.06.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 06/02/2014] [Accepted: 06/17/2014] [Indexed: 11/20/2022]
Abstract
OBJECTIVE Previous studies have shown ezetimibe treatment results in a 2-6-fold increase in reverse cholesterol transport (RCT). However, recent sterol balance studies question the role of biliary sterol secretion in RCT, and challenge the hypothesis that ezetimibe increases RCT through decreased absorption of biliary cholesterol in the intestine. We set out to determine whether ezetimibe may increase RCT by mechanisms that are independent of its well-established inhibition of intestinal cholesterol absorption. METHODS C57BL/6J, Npc1l1-KO, and/or Abcg8-KO mice were fed a chow diet with or without ezetimibe and fecal [(14)C]-neutral and [(14)C]-acidic sterols were measured to examine macrophage-to-feces RCT. We measured the expression of RCT related genes in the liver and jejunum in these mice. To confirm our significant gene expression findings, we utilized primary human hepatocytes cultured with or without a glucuronated metabolite of ezetimibe. RESULTS Our studies revealed that treatment with ezetimibe was associated with increased expression of hepatic Abcg5 and Abcg8. Ezetimibe did not directly affect expression in the liver, but this expression was due to the inhibition of intestinal cholesterol absorption. This conclusion was supported by the absence of an ABCG5/ABCG8 expression response to treatment with an ezetimibe metabolite in primary human hepatocytes. Finally, we found that the ezetimibe-dependent stimulation of RCT was attenuated in the absence of Abcg8. CONCLUSIONS Our study is the first to demonstrate ezetimibe treatment cooperatively stimulated macrophage-to-feces RCT by indirectly increasing liver Abcg5/Abcg8 expression in addition to its known suppression of intestinal cholesterol absorption.
Collapse
Affiliation(s)
- Jessica B Altemus
- Genomic Medicine Institute, The Cleveland Clinic Foundation, Cleveland, OH, USA.
| | - Shailendra B Patel
- Division of Endocrinology, Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI, USA; Division of Endocrinology, Metabolism and Clinical Nutrition, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ephraim Sehayek
- Genomic Medicine Institute, The Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
29
|
Liu X, Hao JJ, Zhang LJ, Zhao X, He XX, Li MM, Zhao XL, Wu JD, Qiu PJ, Yu GL. Activated AMPK explains hypolipidemic effects of sulfated low molecular weight guluronate on HepG2 cells. Eur J Med Chem 2014; 85:304-10. [DOI: 10.1016/j.ejmech.2014.07.107] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 07/28/2014] [Accepted: 07/29/2014] [Indexed: 11/28/2022]
|
30
|
Prashanth A, Jeyakumar SM, Giridharan NV, Vajreswari A. Vitamin A-enriched diet modulates reverse cholesterol transport in hypercholesterolemic obese rats of the WNIN/Ob strain. J Atheroscler Thromb 2014; 21:1197-207. [PMID: 25100235 DOI: 10.5551/jat.22186] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM Vitamin A plays a major role in lipid metabolism. Previously, we reported that chronic vitamin A feeding (129 mg/kg) for two months normalized the abnormally high plasma HDL-cholesterol (HDL-C) levels in hypercholesterolemic obese rats by upregulating the hepatic scavenger receptor class B type 1 (SR-BI) expression. In this report, we hypothesize that the administration of a dose less than 129 mg of vitamin A/kg would also be effective in lowering the plasma HDL-C levels in these rats. METHODS Changes in the activity and expression of proteins related to RCT were analyzed together with blood parameters in five-month-old male lean and obese rats supplemented with 2.6 (control group), 26, 52 and 129 mg of vitamin A/kg as retinyl palmitate for 20 weeks. RESULTS Vitamin A supplementation in the obese rats decreased the plasma HDL-C levels with a concomitant increase in the hepatic SR-BI expression and lipase activity compared to that observed in the control diet-fed obese rats treated with 2.6 mg of vitamin A/kg diet. Furthermore, vitamin A supplementation at doses of 52 and 129 mg/kg diet reduced the plasma lecithin cholesterol acyltransferase activity and increased the hepatic ATP-binding cassette transporter protein A1 expression in the obese rats. Interestingly, most of these changes were not observed in the lean rats fed a vitamin A-enriched diet. CONCLUSIONS Chronic feeding of a vitamin A-enriched diet in hypercholesterolemic obese rats normalizes the plasma HDL-C level and presumably improves RCT, with an effective dose of 52 mg/kg diet. Further studies should focus on the pharmacological potential of vitamin A supplementation to correct an abnormal human obesity-associated lipoprotein metabolism.
Collapse
|
31
|
Yang Y, Li W, Liu Y, Sun Y, Li Y, Yao Q, Li J, Zhang Q, Gao Y, Gao L, Zhao J. Alpha-lipoic acid improves high-fat diet-induced hepatic steatosis by modulating the transcription factors SREBP-1, FoxO1 and Nrf2 via the SIRT1/LKB1/AMPK pathway. J Nutr Biochem 2014; 25:1207-1217. [PMID: 25123628 DOI: 10.1016/j.jnutbio.2014.06.001] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 04/27/2014] [Accepted: 06/01/2014] [Indexed: 02/07/2023]
Abstract
Understanding the mechanism by which alpha-lipoic acid supplementation has a protective effect upon nonalcoholic fatty liver disease in vivo and in vitro may lead to targets for preventing hepatic steatosis. Male C57BL/6J mice were fed a normal diet, high-fat diet or high-fat diet supplemented with alpha-lipoic acid for 24 weeks. HepG2 cells were incubated with normal medium, palmitate or alpha-lipoic acid. The lipid-lowering effects were measured. The protein expression and distribution were analyzed by Western blot, immunoprecipitation and immunofluorescence, respectively. We found that alpha-lipoic acid enhanced sirtuin 1 deacetylase activity through liver kinase B1 and stimulated AMP-activated protein kinase. By activating the sirtuin 1/liver kinase B1/AMP-activated protein kinase pathway, the translocation of sterol regulatory element-binding protein-1 into the nucleus and forkhead box O1 into the cytoplasm was prevented. Alpha-lipoic acid increased adipose triacylglycerol lipase expression and decreased fatty acid synthase abundance. In in vivo and in vitro studies, alpha-lipoic acid also increased nuclear NF-E2-related factor 2 levels and downstream target amounts via the sirtuin 1 pathway. Alpha-lipoic acid eventually reduced intrahepatic and serum triglyceride content. The protective effects of alpha-lipoic acid on hepatic steatosis appear to be associated with the transcription factors sterol regulatory element-binding protein-1, forkhead box O1 and NF-E2-related factor 2.
Collapse
Affiliation(s)
- Yi Yang
- Department of Endocrinology, Provincial Hospital Affiliated to Shandong University, Jinan 250021, China; Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China.
| | - Wang Li
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Yang Liu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Yuning Sun
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Yan Li
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Qing Yao
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Jianning Li
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Qian Zhang
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Yujing Gao
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Ling Gao
- Central Lab. Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Jiajun Zhao
- Department of Endocrinology, Provincial Hospital Affiliated to Shandong University, Jinan 250021, China; Institute of Endocrinology, Shandong Academy of Clinical Medicine, Jinan 250021, China.
| |
Collapse
|
32
|
Vincent J. Reducing elevated plasma LDL cholesterol: the central role of the LDL receptor. Clin Pharmacol Ther 2014; 96:3-7. [PMID: 24942393 DOI: 10.1038/clpt.2014.95] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Elevated low-density lipoprotein cholesterol (LDL-C) is an established risk factor for cardiovascular disease (CVD), and reduction of elevated LDL-C reduces mortality in patients at risk. This benefit has evolved from the use of statins and knowledge of the LDL receptor (LDLR). The most potent drugs used for dyslipidemias act by mechanisms that involve this receptor. Advances in molecular genetics and understanding of the regulation of this receptor have revealed several pharmacological targets that are being explored to develop more targeted therapies for dyslipidemias.
Collapse
Affiliation(s)
- J Vincent
- Pfizer Global Pharmaceuticals, New York, New York, USA
| |
Collapse
|
33
|
Singh AB, Kan CFK, Shende V, Dong B, Liu J. A novel posttranscriptional mechanism for dietary cholesterol-mediated suppression of liver LDL receptor expression. J Lipid Res 2014; 55:1397-407. [PMID: 24792925 DOI: 10.1194/jlr.m049429] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Indexed: 12/21/2022] Open
Abstract
It is well-established that over-accumulation of dietary cholesterol in the liver inhibits sterol-regulatory element binding protein (SREBP)-mediated LDL receptor (LDLR) gene transcription leading to a reduced hepatic LDLR mRNA level in hypercholesterolemic animals. However, it is unknown whether elevated cholesterol levels can elicit a cellular response to increase LDLR mRNA turnover to further repress LDLR expression in liver tissue. In the current study, we examined the effect of a high cholesterol diet on the hepatic expression of LDLR mRNA binding proteins in three different animal models and in cultured hepatic cells. Our results demonstrate that high cholesterol feeding specifically elevates the hepatic expression of LDLR mRNA decay promoting factor heterogeneous nuclear ribonucleoprotein (HNRNP)D without affecting expressions of other LDLR mRNA binding proteins in vivo and in vitro. Employing the approach of adenovirus-mediated gene knockdown, we further show that depletion of HNRNPD in the liver results in a marked reduction of serum LDL-cholesterol and a substantial increase in liver LDLR expression in hyperlipidemic mice. Additional studies of gene knockdown in albumin-luciferase-untranslated region (UTR) transgenic mice provide strong evidence supporting the essential role of 3'UTR in HNRNPD-mediated LDLR mRNA degradation in liver tissue. Altogether, this work identifies a novel posttranscriptional regulatory mechanism by which dietary cholesterol inhibits liver LDLR expression via inducing HNRNPD to accelerate LDLR mRNA degradation.
Collapse
Affiliation(s)
- Amar Bahadur Singh
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 Department of Medicine, Stanford University, Stanford, CA 94305
| | | | - Vikram Shende
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 Department of Medicine, Stanford University, Stanford, CA 94305
| | - Bin Dong
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304
| | - Jingwen Liu
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304
| |
Collapse
|
34
|
Han SI, Komatsu Y, Murayama A, Steffensen KR, Nakagawa Y, Nakajima Y, Suzuki M, Oie S, Parini P, Vedin LL, Kishimoto H, Shimano H, Gustafsson JÅ, Yanagisawa J. Estrogen receptor ligands ameliorate fatty liver through a nonclassical estrogen receptor/Liver X receptor pathway in mice. Hepatology 2014; 59:1791-802. [PMID: 24277692 DOI: 10.1002/hep.26951] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 11/19/2013] [Indexed: 12/17/2022]
Abstract
UNLABELLED Liver X receptor (LXR) activation stimulates triglyceride (TG) accumulation in the liver. Several lines of evidence indicate that estradiol-17β (E2) reduces TG levels in the liver; however, the molecular mechanism underlying the E2 effect remains unclear. Here, we show that administration of E2 attenuated sterol regulatory element-binding protein (SREBP)-1 expression and TG accumulation induced by LXR activation in mouse liver. In estrogen receptor alpha (ERα) knockout (KO) and liver-specific ERα KO mice, E2 did not affect SREBP-1 expression or TG levels. Molecular analysis revealed that ERα is recruited to the SREBP-1c promoter through direct binding to LXR and inhibits coactivator recruitment to LXR in an E2-dependent manner. Our findings demonstrate the existence of a novel liver-dependent mechanism controlling TG accumulation through the nonclassical ER/LXR pathway. To confirm that a nonclassical ER/LXR pathway regulates ERα-dependent inhibition of LXR activation, we screened ERα ligands that were able to repress LXR activation without enhancing ERα transcriptional activity, and, as a result, we identified the phytoestrogen, phloretin. In mice, phloretin showed no estrogenic activity; however, it did reduce SREBP-1 expression and TG levels in liver of mice fed a high-fat diet to an extent similar to that of E2. CONCLUSION We propose that ER ligands reduce TG levels in the liver by inhibiting LXR activation through a nonclassical pathway. Our results also indicate that the effects of ER on TG accumulation can be distinguished from its estrogenic effects by a specific ER ligand.
Collapse
Affiliation(s)
- Song-iee Han
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Nakakuki M, Kawano H, Notsu T, Imada K, Mizuguchi K, Shimano H. A novel processing system of sterol regulatory element-binding protein-1c regulated by polyunsaturated fatty acid. J Biochem 2014; 155:301-13. [PMID: 24729033 DOI: 10.1093/jb/mvu019] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The proteolytic cascade is the key step in transactivation of sterol regulatory element-binding proteins (SREBPs), a transcriptional factor of lipid synthesis. Proteolysis of SREBP-2 is strictly regulated by sterols, but that of SREBP-1c was not strongly sterol-regulated, but inhibited by polyunsaturated fatty acids (PUFAs). In this study, the proteolytic processing of SREBP-1 and -2 was examined by transfection studies of cDNA-encoding mutants in which all the known cleavage sites were disrupted. In cultured cells, sterol-regulated SREBP-2 processing was completely eliminated by mutation of cleavage sites. In contrast, the corresponding SREBP-1c mutants as well as wild type exhibited large amounts of cleaved products in the nuclear extracts from culture cells and murine liver in vivo. The nuclear form of the mutant SREBP-1c was induced by delipidated condition and suppressed by eicosapentaenoic acid, an n-3 PUFA, but not by sterols. This novel processing mechanism was affected by neither SREBP cleavage-activating protein (SCAP) nor insulin-induced gene (Insig)-1, unlike SREBP-2, but abolished by a serine protease inhibitor. Through analysis of deletion mutant, a site-2 protease recognition sequence (DRSR) was identified to be involved in this novel processing. These findings suggest that SREBP-1c cleavage could be subjected to a novel PUFA-regulated cleavage system in addition to the sterol-regulatory SCAP/Insig system.
Collapse
Affiliation(s)
- Masanori Nakakuki
- Development Research, Pharmaceutical Research Center, Mochida Pharmaceutical Co., Ltd., 722 Jimba-aza-Uenohara, Gotemba, Shizuoka 412-8524; and Department of Internal Medicine (Endocrinology and Metabolism), Graduate School of Comprehensive Human Sciences, Faculty of Medicine, and International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | | | | | | | | | | |
Collapse
|
36
|
He S, Hao J, Peng W, Qiu P, Li C, Guan H. Modulation of lipid metabolism by deep-sea water in cultured human liver (HepG2) cells. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2014; 16:219-229. [PMID: 24057172 DOI: 10.1007/s10126-013-9540-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 09/07/2013] [Indexed: 06/02/2023]
Abstract
It has been found that deep-sea water was associated with lower serum lipid in animal model studies. Herein, we investigated whether DSW exerted a hypolipidemic activity and further elucidated how DSW modulated lipid metabolism in HepG2 cells. Preliminary animal studies showed that DSW exhibited potency to decrease serum total cholesterol, triglycerides, and LDL cholesterol, and increase HDL cholesterol, and the hepatic lipid contents were also significantly lower in the DSW group. When DSW was added to HepG2 cells, it decreased the lipid contents of hepatocyte through the activation of AMP-activated protein kinase, thus inhibiting the synthesis of cholesterol and fatty acid. Besides, LDL receptor was upregulated by activation of sterol regulatory element-binding protein-2. In addition, the levels of apolipoprotein AI and cholesterol 7-alpha-hydroxylase were also raised. Our investigation provided mechanisms by which DSW modulated lipid metabolism and indicated that DSW was worthy of further investigation and could be developed as functional drinking water in the prevention and treatment of hypolipidemic and other lifestyle-related diseases.
Collapse
Affiliation(s)
- Shan He
- Key Laboratory of Marine Drugs, Ministry of Education, Marine Drugs and Food Institute, Ocean University of China, Qingdao, 266003, People's Republic of China
| | | | | | | | | | | |
Collapse
|
37
|
Rauer C, Ringseis R, Rothe S, Wen G, Eder K. Sterol regulatory element-binding proteins are regulators of the rat thyroid peroxidase gene in thyroid cells. PLoS One 2014; 9:e91265. [PMID: 24625548 PMCID: PMC3953333 DOI: 10.1371/journal.pone.0091265] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 02/07/2014] [Indexed: 01/21/2023] Open
Abstract
Sterol regulatory element-binding proteins (SREBPs)-1c and -2, which were initially discovered as master transcriptional regulators of lipid biosynthesis and uptake, were recently identified as novel transcriptional regulators of the sodium-iodide symporter gene in the thyroid, which is essential for thyroid hormone synthesis. Based on this observation that SREBPs play a role for thyroid hormone synthesis, we hypothesized that another gene involved in thyroid hormone synthesis, the thyroid peroxidase (TPO) gene, is also a target of SREBP-1c and -2. Thyroid epithelial cells treated with 25-hydroxycholesterol, which is known to inhibit SREBP activation, had about 50% decreased mRNA levels of TPO. Similarly, the mRNA level of TPO was reduced by about 50% in response to siRNA mediated knockdown of both, SREBP-1 and SREBP-2. Reporter gene assays revealed that overexpression of active SREBP-1c and -2 causes a strong transcriptional activation of the rat TPO gene, which was localized to an approximately 80 bp region in the intron 1 of the rat TPO gene. In vitro- and in vivo-binding of both, SREBP-1c and SREBP-2, to this region in the rat TPO gene could be demonstrated using gel-shift assays and chromatin immunoprecipitation. Mutation analysis of the 80 bp region of rat TPO intron 1 revealed two isolated and two overlapping SREBP-binding elements from which one, the overlapping SRE+609/InvSRE+614, was shown to be functional in reporter gene assays. In connection with recent findings that the rat NIS gene is also a SREBP target gene in the thyroid, the present findings suggest that SREBPs may be possible novel targets for pharmacological modulation of thyroid hormone synthesis.
Collapse
Affiliation(s)
- Christine Rauer
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-Universität Gießen, Gießen, Germany
| | - Robert Ringseis
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-Universität Gießen, Gießen, Germany
- * E-mail:
| | - Susanne Rothe
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-Universität Gießen, Gießen, Germany
| | - Gaiping Wen
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-Universität Gießen, Gießen, Germany
| | - Klaus Eder
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-Universität Gießen, Gießen, Germany
| |
Collapse
|
38
|
Miao J, Haas JT, Manthena P, Wang Y, Zhao E, Vaitheesvaran B, Kurland IJ, Biddinger SB. Hepatic insulin receptor deficiency impairs the SREBP-2 response to feeding and statins. J Lipid Res 2014; 55:659-67. [PMID: 24516236 DOI: 10.1194/jlr.m043711] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The liver plays a central role in metabolism and mediating insulin action. To dissect the effects of insulin on the liver in vivo, we have studied liver insulin receptor knockout (LIRKO) mice. Because LIRKO livers lack insulin receptors, they are unable to respond to insulin. Surprisingly, the most profound derangement observed in LIRKO livers by microarray analysis is a suppression of the cholesterologenic genes. Sterol regulatory element binding protein (SREBP)-2 promotes cholesterologenic gene transcription, and is inhibited by intracellular cholesterol. LIRKO livers show a slight increase in hepatic cholesterol, a 40% decrease in Srebp-2, and a 50-90% decrease in the cholesterologenic genes at the mRNA and protein levels. In control mice, SREBP-2 and cholesterologenic gene expression are suppressed by fasting and restored by refeeding; in LIRKO mice, this response is abolished. Similarly, the ability of statins to induce Srebp-2 and the cholesterologenic genes is lost in LIRKO livers. In contrast, ezetimibe treatment robustly induces Srepb-2 and its targets in LIRKO livers, raising the possibility that insulin may regulate SREBP-2 indirectly, by altering the accumulation or distribution of cholesterol within the hepatocyte. Taken together, these data indicate that cholesterol synthesis is a key target of insulin action in the liver.
Collapse
Affiliation(s)
- Ji Miao
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston MA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Qiu XM, Jin CT, Wang W. Association between single nucleotide polymorphisms of sterol regulatory element binding protein-2 gene and risk of knee osteoarthritis in a Chinese Han population. J Int Med Res 2014; 42:320-8. [PMID: 24496149 DOI: 10.1177/0300060513507392] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE To investigate associations between single nucleotide polymorphisms (SNPs) rs2228314 and rs2267443 in the sterol regulatory element binding protein-2 gene (SREBP-2) and knee osteoarthritis (OA) susceptibility in a Chinese Han population. METHODS SREBP-2 rs2228314 and rs2267443 polymorphisms were genotyped in patients with knee OA and age- and sex-matched OA-free controls from a Chinese Han population. RESULTS A total of 402 patients with knee OA and 410 controls were enrolled in the study. GC and CC genotypes of rs2228314, and variant C, were associated with a significantly increased risk of knee OA. On stratification analysis, the association between the risk of OA and rs2228314 GC heterozygotes compared with GG homozygotes was stronger in females and those aged >65 years. In contrast, the GA and AA genotypes of rs2267443 were not significantly associated with the risk of knee OA, even after further stratification analysis according to age or sex. CONCLUSIONS SREBP-2 rs2228314 G to C change and variant C genotype may contribute to knee OA risk in a Chinese Han population.
Collapse
Affiliation(s)
- Xiao-Ming Qiu
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | | |
Collapse
|
40
|
Tanaka S, Yamazaki T, Asano S, Mitsumoto A, Kobayashi D, Kudo N, Kawashima Y. Increased lipid synthesis and decreased β-oxidation in the liver of SHR/NDmcr-cp (cp/cp) rats, an animal model of metabolic syndrome. Lipids 2013; 48:1115-34. [PMID: 24045975 DOI: 10.1007/s11745-013-3839-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 08/24/2013] [Indexed: 12/19/2022]
Abstract
SHR/NDmcr-cp (cp/cp) rats (SHR/NDcp) are an animal model of metabolic syndrome. A previous study of ours revealed drastic increases in the mass of palmitic (16:0), oleic (18:1n-9), palmitoleic (16:1n-7), cis-vaccenic (18:1n-7) and 5,8,11-eicosatrienoic acids in the liver of SHR/NDcp. However, detailed information on the class of lipid accumulated and the mechanism responsible for the overproduction of the accumulated lipid in the liver was not obtained. This study aimed to characterize the class of lipid accumulated and to explore the mechanism underlying the lipid accumulation in the liver of SHR/NDcp, in comparison with SHR/NDmcr-cp (+/+) (lean hypertensive littermates of SHR/NDcp) and Wistar Kyoto rats. In the liver of SHR/NDcp, de novo synthesis of fatty acids (16:0, 18:1n-9 and 16:1n-7) and triacylglycerol (TAG) synthesis were up-regulated and fatty acid β-oxidation was down-regulated. These perturbations of lipid metabolism caused fat accumulation in hepatocytes and accumulation of TAG, which were enriched with 16:0, 18:1n-9 and 16:1n-7, in the liver of SHR/NDcp. On the other hand, no changes were found in hepatic contents of diacylglycerol and unesterified fatty acid (FFA); among FFA, there were no differences in the hepatic concentrations of unesterified 16:0 and stearic acid between SHR/NDcp and two other groups of rats. Moreover, little change was brought about in the expression of genes responsive to endoplasmic reticulum stress in the liver of SHR/NDcp. These results may reinforce the pathophysiological role of stearoyl-CoA desaturase 1 and fatty acid elongase 6 in the liver of SHR/NDcp.
Collapse
Affiliation(s)
- Shizuyo Tanaka
- Faculty of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | | | | | | | | | | | | |
Collapse
|
41
|
Fukuda N, Ito T, Ohwada K, Fujii J. Upregulation of fatty acid synthesis and the suppression of hepatic triglyceride lipase as a direct cause of hereditary postprandial hypertriglyceridemia in rabbits. J Clin Biochem Nutr 2013; 53:114-21. [PMID: 24062609 PMCID: PMC3774926 DOI: 10.3164/jcbn.13-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 06/16/2013] [Indexed: 12/03/2022] Open
Abstract
Rabbits with hereditary postprandial hypertriglyceridemia exhibit central obesity and are regarded as a reliable model for metabolic syndrome. This study was performed to gain insight into the affected process of lipid metabolism and into the causative genes of the postprandial hypertriglyceridemia rabbits. Eleven genes that play key roles in lipid metabolism were selected, their mRNA levels were assessed by quantitative PCR, and their expressions were compared among postprandial hypertriglyceridemia rabbits using Japanese white rabbits as the control. Two genes appeared to be in causal connection with postprandial hypertriglyceridemia, and these were regarded as likely candidates for the pathogenesis. One was the fatty acid synthase gene, which had an expression constitutively higher in postprandial hypertriglyceridemia rabbits than in Japanese white rabbits during the fasting state and reached quite high levels after feeding. The other was the gene for hepatic triglyceride lipase with an expression that was approximately one order lower than that found in the Japanese white rabbits. The low plasma hepatic triglyceride lipase activities were consistent with the low levels of the transcript in the livers of the postprandial hypertriglyceridemia rabbits. Thus, elevated fatty acid synthesis and defected lipid hydrolysis together would cause the postprandial hypertriglyceridemia in postprandial hypertriglyceridemia rabbits.
Collapse
Affiliation(s)
- Naoki Fukuda
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-9585, Japan
| | | | | | | |
Collapse
|
42
|
Jeon TI, Esquejo RM, Roqueta-Rivera M, Phelan PE, Moon YA, Govindarajan SS, Esau CC, Osborne TF. An SREBP-responsive microRNA operon contributes to a regulatory loop for intracellular lipid homeostasis. Cell Metab 2013; 18:51-61. [PMID: 23823476 PMCID: PMC3740797 DOI: 10.1016/j.cmet.2013.06.010] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 03/13/2013] [Accepted: 06/12/2013] [Indexed: 12/11/2022]
Abstract
Sterol regulatory element-binding proteins (SREBPs) have evolved as a focal point for linking lipid synthesis with other pathways that regulate cell growth and survival. Here, we have uncovered a polycistrionic microRNA (miRNA) locus that is activated directly by SREBP-2. Two of the encoded miRNAs, miR-182 and miR-96, negatively regulate the expression of Fbxw7 and Insig-2, respectively, and both are known to negatively affect nuclear SREBP accumulation. Direct manipulation of this miRNA pathway alters nuclear SREBP levels and endogenous lipid synthesis. Thus, we have uncovered a mechanism for the regulation of intracellular lipid metabolism mediated by the concerted action of a pair of miRNAs that are expressed from the same SREBP-2-regulated miRNA locus, and each targets a different protein of the multistep pathway that regulates SREBP function. These studies reveal an miRNA "operon" analogous to the classic model for genetic control in bacterial regulatory systems.
Collapse
Affiliation(s)
- Tae-Il Jeon
- Metabolic Signaling and Disease Program and Diabetes and Obesity Center, Sanford-Burnham Medical Research Institute, Orlando, Florida 32827, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Lee SD, Thornton SJ, Sachs-Barrable K, Kim JH, Wasan KM. Evaluation of the contribution of the ATP binding cassette transporter, P-glycoprotein, to in vivo cholesterol homeostasis. Mol Pharm 2013; 10:3203-12. [PMID: 23750858 DOI: 10.1021/mp4002415] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
P-glycoprotein (Pgp, encoded by ABCB1, commonly known as MDR1), an ATP-dependent transporter with a broad range of hydrophobic drug substrates, has been associated with the in vitro intracellular transport of cholesterol; however, these findings have not been confirmed in vivo. In this manuscript we tested the contributions of Pgp to in vivo cholesterol homeostasis by comparing the cholesterol phenotype of wild type mice with mice lacking both murine isoforms of Pgp (Abcb1a(-/-)/1b(-/-)) by measuring cholesterol absorption, circulating cholesterol, and lipoprotein cholesterol profiles. The mice were fed diets containing normal or high levels of dietary fat (25% vs 45% kcal from fat) and cholesterol (0.02% vs 0.20% w/w) for 8 weeks to challenge their capacity to maintain homeostasis. There were no significant differences in cholesterol absorption, circulating cholesterol levels, and lipoprotein profiles between Pgp knockout and wild type mice fed matching diets. Compensatory shifts were observed in the activation of two key transcription factors involved in maintaining cholesterol balance, the Liver X Receptor and SREBP-2, which may have maintained the wild type phenotype in the knockout mice. Deletion of Pgp affected the molar composition of gallbladder bile, when the mice were fed diets containing high levels of dietary fat, cholesterol, or both. The mole fraction of bile salts was reduced in the gallbladder bile of Pgp knockout mice, while the mole fraction of cholesterol was increased. In this paper, we provide evidence that Pgp knockout mice maintain cholesterol homeostasis, even when challenged with high cholesterol diets. We suggest that the specific shifts in cholesterol regulatory networks identified in the jejunum and liver of the knockout mice may have compensated for the lack of Pgp. Our finding that Pgp knockout mice were unable to maintain gallbladder bile composition when challenged with high dietary fat and/or cholesterol compliments recent reports that Pgp may be a secondary bile salt export pump.
Collapse
Affiliation(s)
- Stephen D Lee
- Faculty of Pharmaceutical Sciences, University of British Columbia , 2405 Wesbrook Mall, Vancouver, BC, Canada V6T 1Z3
| | | | | | | | | |
Collapse
|
44
|
Lagace TA, Ridgway ND. The role of phospholipids in the biological activity and structure of the endoplasmic reticulum. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:2499-510. [PMID: 23711956 DOI: 10.1016/j.bbamcr.2013.05.018] [Citation(s) in RCA: 170] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 05/09/2013] [Accepted: 05/15/2013] [Indexed: 01/22/2023]
Abstract
The endoplasmic reticulum (ER) is an interconnected network of tubular and planar membranes that supports the synthesis and export of proteins, carbohydrates and lipids. Phospholipids, in particular phosphatidylcholine (PC), are synthesized in the ER where they have essential functions including provision of membranes required for protein synthesis and export, cholesterol homeostasis, and triacylglycerol storage and secretion. Coordination of these biological processes is essential, as highlighted by findings that link phospholipid metabolism in the ER with perturbations in lipid storage/secretion and stress responses, ultimately contributing to obesity/diabetes, atherosclerosis and neurological disorders. Phospholipid synthesis is not uniformly distributed in the ER but is localized at membrane interfaces or contact zones with other organelles, and in dynamic, proliferating ER membranes. The topology of phospholipid synthesis is an important consideration when establishing the etiology of diseases that arise from ER dysfunction. This review will highlight our current understanding of the contribution of phospholipid synthesis to proper ER function, and how alterations contribute to aberrant stress responses and disease. This article is part of a Special Issue entitled: Functional and structural diversity of endoplasmic reticulum.
Collapse
Affiliation(s)
- Thomas A Lagace
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada.
| | | |
Collapse
|
45
|
Ignatova ID, Angdisen J, Moran E, Schulman IG. Differential regulation of gene expression by LXRs in response to macrophage cholesterol loading. Mol Endocrinol 2013; 27:1036-47. [PMID: 23686114 DOI: 10.1210/me.2013-1051] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The ability of cells to precisely control gene expression in response to intracellular and extracellular signals plays an important role in both normal physiology and in pathological settings. For instance, the accumulation of excess cholesterol by macrophages initiates a genetic response mediated by the liver X receptors (LXRs)-α (NR1H3) and LXRβ (NR1H2), which facilitates the transport of cholesterol out of cells to high-density lipoprotein particles. Studies using synthetic LXR agonists have also demonstrated that macrophage LXR activation simultaneously induces a second network of genes that promotes fatty acid and triglyceride synthesis that may support the detoxification of excess free cholesterol by storage in the ester form. We now show that treatment of human THP-1 macrophages with endogenous or synthetic LXR ligands stimulates both transcriptional and posttranscriptional pathways that result in the selective recruitment of the LXRα subtype to LXR-regulated promoters. Interestingly, when human or mouse macrophages are loaded with cholesterol under conditions that mimic the development of atherogenic macrophage foam cells, a selective LXR response is generated that induces genes mediating cholesterol transport but does not coordinately regulate genes involved in fatty acid synthesis. The gene-selective response to cholesterol loading occurs, even in the presence of LXRα binding to the promoter of the gene encoding the sterol regulatory element-binding protein-1c, the master transcriptional regulator of fatty acid synthesis. The ability of promoter bound LXRα to recruit RNA polymerase to the sterol regulatory element-binding protein-1c promoter, however, appears to be ligand selective.
Collapse
Affiliation(s)
- Irena D Ignatova
- Department of Pharmacology, University of Virginia, 1300 Jefferson Park Avenue, PO Box 800735, Charlottesville, Virginia 22908, USA.
| | | | | | | |
Collapse
|
46
|
Monacolin K affects lipid metabolism through SIRT1/AMPK pathway in HepG2 cells. Arch Pharm Res 2013; 36:1541-51. [PMID: 23657807 DOI: 10.1007/s12272-013-0150-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 05/02/2013] [Indexed: 10/26/2022]
Abstract
Monacolin K is the secondary metabolite isolated from Monascus spp. It is the natural form of lovastatin, which is clinically used to reduce the synthesis of cholesterol by inhibiting 3-hydroxy-3-methylglutaryl coenzyme A reductase. In the present study, monacolin K increased protein expression of SIRT1 and phosphorylation level of AMP-activated protein kinase (AMPK) in HepG2 cells. Through activation of SIRT1/AMPK pathway, monacolin K increased phosphorylation of acetyl CoA carboxylase and caused nuclear translocation of forkhead box O1. The western blotting results showed that monacolin K increased expression of adipose triglyceride lipase but decreased abundances of fatty acid synthase (FAS) and sterol regulatory element-binding protein 1 (SREBP1). Monacolin K also decreased the intracellular accumulation of lipids as demonstrated by Oil Red O staining. In addition, the immunostaining showed that monacolin K prevented the nuclear translocation of SREBP1, indicating the association with down-regulation of FAS. All the demonstrated effects of monacolin K were counteracted by nicotinamide or compound C, the inhibitors of SIRT1 or AMPK. In summary, monacolin K reduces the lipid content through SIRT1/AMPK pathway in HepG2 cells, which promotes catabolism and inhibits anabolism of lipid.
Collapse
|
47
|
Kumazaki M, Ando H, Ushijima K, Fujimura A. Comparative effects of statins on murine cardiac gene expression profiles in normal mice. Eur J Pharmacol 2013; 707:71-7. [DOI: 10.1016/j.ejphar.2013.03.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 03/12/2013] [Accepted: 03/13/2013] [Indexed: 12/01/2022]
|
48
|
Oh YS, Lee YJ, Kang Y, Han J, Lim OK, Jun HS. Exendin-4 inhibits glucolipotoxic ER stress in pancreatic β cells via regulation of SREBP1c and C/EBPβ transcription factors. J Endocrinol 2013; 216:343-52. [PMID: 23257266 DOI: 10.1530/joe-12-0311] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Prolonged exposure to high glucose (HG) and palmitate (PA) results in increased ER stress and subsequently induces β-cell apoptosis. Exendin-4, a glucagon-like peptide-1 agonist, is known to protect β cells from toxicity induced by cytokines, HG, or fatty acids by reducing ER stress. However, the detailed molecular mechanisms for this protective effect are still not known. In this study, we investigated the role of exendin-4 in the inhibition of glucolipotoxicity-induced ER stress and β-cell apoptosis. Exendin-4 treatment protected INS-1 β cells from apoptosis in response to HG/PA (25 mM glucose+400 μM PA). HG/PA treatment increased cleaved caspase-3 and induced ER stress maker proteins such as PERK (EIF2AK3), ATF6, and phosphorylated forms of PERK, eIF2α, IRE1α (ERN1), and JNK (MAPK8), and these increases were significantly inhibited by exendin-4 treatment. HG/PA treatment of INS-1 cells increased SREBP1 (SREBF1) protein and induced its nuclear translocation and subsequently increased C/EBPβ (CEBPB) protein and its nuclear translocation. Exendin-4 treatment attenuated this increase. Knockdown of SREBP1c reduced the activation of C/EBPβ and also blocked the expression of ER stress markers induced by HG/PA treatment. Our results indicate that exendin-4 inhibits the activation of SREBP1c and C/EBPβ, which, in turn, may reduce glucolipotoxicity-induced ER stress and β-cell apoptosis.
Collapse
Affiliation(s)
- Yoon Sin Oh
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 7-45 Songdo-dong, Yeonsu-ku, Incheon, Korea
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
Dietary fat is well recognised as an important macronutrient that has major effects on growth, development and health of all animals including humans. The amount and type of fat in the diet impacts on many aspects of metabolism including lipoprotein pathways, lipid synthesis and oxidation, adipocyte differentiation and cholesterol metabolism. It has become increasingly apparent that many of these effects may be due to direct modulation of expression of key genes through the interaction of fatty acids with certain transcription factors. Peroxisome proliferator-activated receptors (PPARs), the liver X receptors (LXRs), hepatic nuclear factor 4 (HNF-4) and sterol regulatory binding proteins (SREBPs) represent four such factors. This review focuses on emerging evidence that the activity of these transcription factors are regulated by fatty acids and the interactions between them may be responsible for many of the effects of fatty acids on metabolism and the development of chronic disease.
Collapse
|
50
|
Engelking LJ, McFarlane MR, Li CK, Liang G. Blockade of cholesterol absorption by ezetimibe reveals a complex homeostatic network in enterocytes. J Lipid Res 2012; 53:1359-68. [PMID: 22523394 PMCID: PMC3371247 DOI: 10.1194/jlr.m027599] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Indexed: 12/29/2022] Open
Abstract
Enterocyte cholesterol homeostasis reflects aggregated rates of sterol synthesis, efflux, and uptake from plasma and gut lumen. Cholesterol synthesis and LDL uptake are coordinately regulated by sterol regulatory element-binding proteins (SREBP), whereas sterol efflux is regulated by liver X receptors (LXR). How these processes are coordinately regulated in enterocytes, the site of cholesterol absorption, is not well understood. Here, we treat mice with ezetimibe to investigate the effect of blocking cholesterol absorption on intestinal SREBPs, LXRs, and their effectors. Ezetimibe increased nuclear SREBP-2 8-fold. HMG-CoA reductase (HMGR) and LDL receptor (LDLR) mRNA levels increased less than 3-fold, whereas their protein levels increased 30- and 10-fold, respectively. Expression of inducible degrader of LDLR (IDOL), an LXR-regulated gene that degrades LDLRs, was reduced 50% by ezetimibe. Coadministration of ezetimibe with the LXR agonist T0901317 abolished the reduction in IDOL and prevented the increase in LDLR protein. Ezetimibe-stimulated LDLR expression was independent of proprotein convertase subtilisin/kexin type 9 (PSCK9), a protein that degrades LDLRs. To maintain cholesterol homeostasis in the face of ezetimibe, enterocytes boost LDL uptake by increasing LDLR number, and they boost sterol synthesis by increasing HMGR and other cholesterologenic genes. These studies reveal a hitherto undescribed homeostatic network in enterocytes triggered by blockade of cholesterol absorption.
Collapse
Affiliation(s)
- Luke J Engelking
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | | | | | | |
Collapse
|