1
|
Humayrah W, Sabrina N, Stefani M, Taslim NA, Surya R, Handoko MN, Lau V, Hardinsyah H, Tallei TE, Syahputra RA, Nurkolis F. The role of micro-ribonucleic acid and small interfering-ribonucleic acid in precision nutrition for obesity management. Clin Nutr ESPEN 2025; 67:463-475. [PMID: 40158690 DOI: 10.1016/j.clnesp.2025.03.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND & AIMS Precision nutrition aims to tailor dietary interventions based on genetic and molecular profiles. MicroRNAs (miRNAs) and small interfering RNAs (siRNAs) are emerging as critical tools in precision obesity management. miRNAs serve as biomarkers for predicting dietary response and obesity risk, while siRNAs provide a targeted approach to silencing obesity-related genes. This review explores the mechanisms, applications, and potential of integrating miRNA and siRNA in personalized dietary strategies to combat obesity. METHODS A comprehensive literature review was conducted using Boolean operations to identify studies on miRNAs, siRNAs, and their roles in precision nutrition. The review focused on molecular mechanisms, clinical applications, challenges, and future directions in integrating miRNA detection and siRNA therapy for obesity management. RESULTS miRNAs regulate gene expression related to lipid metabolism, adipogenesis, and insulin sensitivity, with miRNA-33 and miRNA-103/107 being notable examples. siRNAs offer precise gene silencing for targets like SREBP-1c and PPARγ, addressing metabolic pathways resistant to dietary interventions. The synergistic integration of miRNAs as biomarkers and siRNAs as therapeutic tools enhances the personalization and efficacy of obesity management. CONCLUSIONS The dual application of miRNAs and siRNAs in precision nutrition represents a transformative approach to obesity management. While challenges such as molecular stability and delivery systems persist, advancements in RNA technology and clinical research promise to revolutionize personalized dietary strategies. Future research should focus on large-scale trials and ethical considerations to ensure equitable and effective implementation.
Collapse
Affiliation(s)
- Wardina Humayrah
- Nutrition Study Program, Faculty of Food Technology and Health, Sahid University, Jakarta, Indonesia.
| | - Nindy Sabrina
- Nutrition Study Program, Faculty of Food Technology and Health, Sahid University, Jakarta, Indonesia.
| | - Megah Stefani
- Nutrition Study Program, Faculty of Food Technology and Health, Sahid University, Jakarta, Indonesia.
| | - Nurpudji Astuti Taslim
- Division of Clinical Nutrition, Department of Nutrition, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia.
| | - Reggie Surya
- Department of Food Technology, Faculty of Engineering, Bina Nusantara University, Jakarta 11480, Indonesia.
| | - Matthew Nathaniel Handoko
- MSc Obesity and Clinical Nutrition, Division of Medicine, Faculty of Medical Siences, University College London, London WC1E 6BT, United Kingdom.
| | - Vincent Lau
- Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia.
| | - Hardinsyah Hardinsyah
- Division of Applied Nutrition, Department of Community Nutrition, Faculty of Human Ecology, IPB University, West Java, Bogor, 16680, Indonesia.
| | - Trina Ekawati Tallei
- Department of Biology, Faculty of Mathematics and Natural Sciences, Sam Ratulangi University, Manado 95115, Indonesia.
| | - Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, University of North Sumatra, Medan 20155, Indonesia.
| | - Fahrul Nurkolis
- Medical Research Center of Indonesia, Surabaya, Indonesia; State Islamic University of Sunan Kalijaga (UIN Sunan Kalijaga), Yogyakarta 55281, Indonesia; Master of Basic Medical Science, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia.
| |
Collapse
|
2
|
Liu CM, Killion EA, Hammoud R, Lu SC, Komorowski R, Liu T, Kanke M, Thomas VA, Cook K, Sivits GN, Ben AB, Atangan LI, Hussien R, Tang A, Shkumatov A, Li CM, Drucker DJ, Véniant MM. GIPR-Ab/GLP-1 peptide-antibody conjugate requires brain GIPR and GLP-1R for additive weight loss in obese mice. Nat Metab 2025:10.1038/s42255-025-01295-w. [PMID: 40301582 DOI: 10.1038/s42255-025-01295-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 03/28/2025] [Indexed: 05/01/2025]
Abstract
Glucose-dependent insulinotropic polypeptide receptor (GIPR) and glucagon-like peptide 1 receptor (GLP-1R) are expressed in the central nervous system (CNS) and regulate food intake. Here, we demonstrate that a peptide-antibody conjugate that blocks GIPR while simultaneously activating GLP-1R (GIPR-Ab/GLP-1) requires both CNS GIPR and CNS GLP-1R for maximal weight loss in obese, primarily male, mice. Moreover, dulaglutide produces greater weight loss in CNS GIPR knockout (KO) mice, and the weight loss achieved with dulaglutide + GIPR-Ab is attenuated in CNS GIPR KO mice. Wild-type mice treated with GIPR-Ab/GLP-1 and CNS GIPR KO mice exhibit similar changes in gene expression related to tissue remodelling, lipid metabolism and inflammation in white adipose tissue and liver. Moreover, GIPR-Ab/GLP-1 is detected in circumventricular organs in the brain and activates c-FOS in downstream neural substrates involved in appetite regulation. Hence, both CNS GIPR and GLP-1R signalling are required for the full weight loss effect of a GIPR-Ab/GLP-1 peptide-antibody conjugate.
Collapse
Affiliation(s)
- Clarissa M Liu
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA
- Amgen R&D Postdoctoral Fellows Program, Amgen Inc., Thousand Oaks, CA, USA
| | - Elizabeth A Killion
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA
| | - Rola Hammoud
- The Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Shu-Chen Lu
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA
| | - Renee Komorowski
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA
| | - Tongyu Liu
- Center for Research Acceleration by Digital Innovation, Amgen Research, Thousand Oaks, CA, USA
| | - Matt Kanke
- Department of Research Technologies, Amgen Research, South San Francisco, CA, USA
| | - Veena A Thomas
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, CA, USA
| | - Kevin Cook
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, CA, USA
| | - Glenn N Sivits
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA
| | - Aerielle B Ben
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA
| | - Larissa I Atangan
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA
| | - Rajaa Hussien
- Department of Translational Safety and Bioanalytical Sciences, Amgen Research, South San Francisco, CA, USA
| | - Amy Tang
- Department of Translational Safety and Bioanalytical Sciences, Amgen Research, South San Francisco, CA, USA
| | - Artem Shkumatov
- Department of Translational Safety and Bioanalytical Sciences, Amgen Research, South San Francisco, CA, USA
| | - Chi-Ming Li
- Department of Research Technologies, Amgen Research, South San Francisco, CA, USA
| | - Daniel J Drucker
- The Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Murielle M Véniant
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA.
| |
Collapse
|
3
|
Canová N, Šípková J, Arora M, Pavlíková Z, Kučera T, Šeda O, Šopin T, Vacík T, Slanař O. Effects of celastrol on the heart and liver galaninergic system expression in a mouse model of Western-type diet-induced obesity and metabolic dysfunction-associated steatotic liver disease and steatohepatitis. Front Pharmacol 2025; 16:1476994. [PMID: 39968178 PMCID: PMC11832397 DOI: 10.3389/fphar.2025.1476994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 01/06/2025] [Indexed: 02/20/2025] Open
Abstract
Background The complexity of the galaninergic system is still not fully understood, especially under specific pre-existing comorbidities related to metabolic dysfunction. A plant-derived triterpenoid celastrol was demonstrated to exert a complex effect on the galaninergic system and to have hepatoprotective and anti-obesity properties. However, the exact molecular mechanisms responsible for these effects remain unclear. Specifically, there are no data on the impact of celastrol on the heart and liver galaninergic system. Therefore, this study aimed to investigate the effects of celastrol on the galaninergic system expression in the heart and liver of mice suffering from diet-induced obesity and metabolic dysfunction-associated steatotic liver disease and steatohepatitis (MASLD/MASH). Methods The male mice C57BL/6J were fed a Western-type high-fat diet for 16 and 20 weeks to induce obesity and MASLD/MASH. Celastrol was administered along with a specific diet for the last 4 weeks to evaluate its impact on the progression of these conditions. Moreover, the inhibitor of sterol regulatory element-binding protein 1/2 (SREBP1/2), fatostatin, was also tested to compare its influence on the galaninergic system with celastrol. Results The study demonstrates that celastrol treatment was safe and led to a reduction in food and energy intake, body fat and liver weights, and MASLD-to-MASH progression and improved glucose tolerance, serum biochemistry markers, and hepatic lipid peroxidation in mice. Quantitative gene expression originally showed significant regulation of galanin and all three of its receptors (GalR1/2/3) in the heart ventricles and only GalR2 in the liver of obese mice. Celastrol influenced the gene expression of galanin receptors: it downregulated Galr1 in the heart and upregulated Galr2 in the liver and Galr3 in the heart ventricles, potentially affecting energy metabolism, oxidative stress, and inflammation. Fatostatin suppressed gene expression of all the detected members of the galaninergic system in the heart ventricles, depicting the role of SREBP in this process. Conclusion These findings suggest that celastrol may beneficially modulate the galaninergic system under obesity and MASLD-to-MASH progression, indicating its potential as a therapeutic agent for disorders associated with metabolic dysfunction.
Collapse
Affiliation(s)
- Nikolina Canová
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Jana Šípková
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Mahak Arora
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Zuzana Pavlíková
- Institute of Histology and Embryology, First Faculty of Medicine, Charles University, Prague, Czechia
- Department of Anthropology and Human Genetics, Faculty of Science, Charles University, Prague, Czechia
| | - Tomáš Kučera
- Institute of Histology and Embryology, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Ondřej Šeda
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Tijana Šopin
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Tomáš Vacík
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Ondřej Slanař
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| |
Collapse
|
4
|
Klag KA, Bell R, Jia X, Seguin A, Maschek JA, Bronner M, Cox JE, Round JL, Ward DM. Low-Iron Diet-Induced Fatty Liver Development Is Microbiota Dependent and Exacerbated by Loss of the Mitochondrial Iron Importer Mitoferrin2. Nutrients 2024; 16:1804. [PMID: 38931165 PMCID: PMC11206261 DOI: 10.3390/nu16121804] [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: 05/01/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Iron deficiency is the number one nutritional problem worldwide. Iron uptake is regulated at the intestine and is highly influenced by the gut microbiome. Blood from the intestines drains directly into the liver, informing iron status and gut microbiota status. Changes in either iron or the microbiome are tightly correlated with the development of metabolic dysfunction-associated steatotic liver disease (MASLD). To investigate the underlying mechanisms of the development of MASLD that connect altered iron metabolism and gut microbiota, we compared specific pathogen free (SPF) or germ-free (GF) mice, fed a normal or low-iron diet. SPF mice on a low-iron diet showed reduced serum triglycerides and MASLD. In contrast, GF low-iron diet-fed mice showed increased serum triglycerides and did not develop hepatic steatosis. SPF mice showed significant changes in liver lipid metabolism and increased insulin resistance that was dependent upon the presence of the gut microbiota. We report that total body loss of mitochondrial iron importer Mitoferrin2 (Mfrn2-/-) exacerbated the development of MASLD on a low-iron diet with significant lipid metabolism alterations. Our study demonstrates a clear contribution of the gut microbiome, dietary iron, and Mfrn2 in the development of MASLD and metabolic syndrome.
Collapse
Affiliation(s)
- Kendra A. Klag
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA; (K.A.K.); (R.B.); (X.J.); (A.S.); (M.B.); (J.L.R.)
| | - Rickesha Bell
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA; (K.A.K.); (R.B.); (X.J.); (A.S.); (M.B.); (J.L.R.)
| | - Xuan Jia
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA; (K.A.K.); (R.B.); (X.J.); (A.S.); (M.B.); (J.L.R.)
| | - Alexandra Seguin
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA; (K.A.K.); (R.B.); (X.J.); (A.S.); (M.B.); (J.L.R.)
| | - J. Alan Maschek
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, UT 84112, USA; (J.A.M.); (J.E.C.)
| | - Mary Bronner
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA; (K.A.K.); (R.B.); (X.J.); (A.S.); (M.B.); (J.L.R.)
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA
| | - James E. Cox
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, UT 84112, USA; (J.A.M.); (J.E.C.)
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - June L. Round
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA; (K.A.K.); (R.B.); (X.J.); (A.S.); (M.B.); (J.L.R.)
| | - Diane M. Ward
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA; (K.A.K.); (R.B.); (X.J.); (A.S.); (M.B.); (J.L.R.)
| |
Collapse
|
5
|
Su F, Koeberle A. Regulation and targeting of SREBP-1 in hepatocellular carcinoma. Cancer Metastasis Rev 2024; 43:673-708. [PMID: 38036934 PMCID: PMC11156753 DOI: 10.1007/s10555-023-10156-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 11/10/2023] [Indexed: 12/02/2023]
Abstract
Hepatocellular carcinoma (HCC) is an increasing burden on global public health and is associated with enhanced lipogenesis, fatty acid uptake, and lipid metabolic reprogramming. De novo lipogenesis is under the control of the transcription factor sterol regulatory element-binding protein 1 (SREBP-1) and essentially contributes to HCC progression. Here, we summarize the current knowledge on the regulation of SREBP-1 isoforms in HCC based on cellular, animal, and clinical data. Specifically, we (i) address the overarching mechanisms for regulating SREBP-1 transcription, proteolytic processing, nuclear stability, and transactivation and (ii) critically discuss their impact on HCC, taking into account (iii) insights from pharmacological approaches. Emphasis is placed on cross-talk with the phosphatidylinositol-3-kinase (PI3K)-protein kinase B (Akt)-mechanistic target of rapamycin (mTOR) axis, AMP-activated protein kinase (AMPK), protein kinase A (PKA), and other kinases that directly phosphorylate SREBP-1; transcription factors, such as liver X receptor (LXR), peroxisome proliferator-activated receptors (PPARs), proliferator-activated receptor γ co-activator 1 (PGC-1), signal transducers and activators of transcription (STATs), and Myc; epigenetic mechanisms; post-translational modifications of SREBP-1; and SREBP-1-regulatory metabolites such as oxysterols and polyunsaturated fatty acids. By carefully scrutinizing the role of SREBP-1 in HCC development, progression, metastasis, and therapy resistance, we shed light on the potential of SREBP-1-targeting strategies in HCC prevention and treatment.
Collapse
Affiliation(s)
- Fengting Su
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Andreas Koeberle
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria.
| |
Collapse
|
6
|
Habelreeh HH, Athinarayanan J, Periasamy VS, Alshatwi AA. Maillard Reaction-Derived S-Doped Carbon Dots Promotes Downregulation of PPARγ, C/EBPα, and SREBP-1 Genes In-Vitro. Molecules 2024; 29:2008. [PMID: 38731499 PMCID: PMC11085050 DOI: 10.3390/molecules29092008] [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: 04/02/2024] [Revised: 04/21/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Carbon nanodots (CDs) are commonly found in food products and have attracted significant attention from food scientists. There is a high probability of CD exposure in humans, but its impacts on health are unclear. Therefore, health effects associated with CD consumption should be investigated. In this study, we attempted to create a model system of the Maillard reaction between cystine and glucose using a simple cooking approach. The CDs (CG-CDs) were isolated from cystine-glucose-based Maillard reaction products and characterized using fluorescence spectroscopy, X-ray diffractometer (XRD), and transmission electron microscope (TEM). Furthermore, human mesenchymal stem cells (hMCs) were used as a model to unravel the CDs' cytotoxic properties. The physiochemical assessment revealed that CG-CDs emit excitation-dependent fluorescence and possess a circular shape with sizes ranging from 2 to 13 nm. CG-CDs are predominantly composed of carbon, oxygen, and sulfur. The results of the cytotoxicity evaluation indicate good biocompatibility, where no severe toxicity was observed in hMCs up to 400 μg/mL. The DPPH assay demonstrated that CDs exert potent antioxidant abilities. The qPCR analysis revealed that CDs promote the downregulation of the key regulatory genes, PPARγ, C/EBPα, SREBP-1, and HMGCR, coupled with the upregulation of anti-inflammatory genes. Our findings suggested that, along with their excellent biocompatibility, CG-CDs may offer positive health outcomes by modulating critical genes involved in lipogenesis, homeostasis, and obesity pathogenesis.
Collapse
Affiliation(s)
| | | | | | - Ali A. Alshatwi
- Nanobiotechnology and Molecular Biology Research Laboratory, Department of Food Science and Nutrition, College of Food Science and Agriculture, King Saud University, P.O. Box 2460, Riyadh 11451, Saudi Arabia; (H.H.H.); (J.A.)
| |
Collapse
|
7
|
Yang D, Shen J, Tang C, Lu Z, Lu F, Bie X, Meng F, Zhao H. Prevention of high-fat-diet-induced obesity in mice by soluble dietary fiber from fermented and unfermented millet bran. Food Res Int 2024; 179:113974. [PMID: 38342528 DOI: 10.1016/j.foodres.2024.113974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/29/2023] [Accepted: 01/03/2024] [Indexed: 02/13/2024]
Abstract
Obesity-related diabetes, cardiovascular disease, and hypertension pose many risks to human health. Thus, mice on a high-fat diet were gavaged with millet bran (unfermented/fermented) soluble dietary fiber (RSDF/FSDF, 500 mg·kg-1) for 10 weeks in current research, and then evaluated the various biological indicators. These findings revealed that RSDF and FSDF supplements could prevent fat synthesis by inhibiting sterol regulatory element-binding protein-1c gene expression. The RSDF supplements can also accelerate fat catabolism through enhanced the mRNA expression levels of adipose triglyceride lipase and peroxisome proliferator-activated receptor α. FSDF supplements can prevent obesity by decreasing 3-hydroxy-3-methyl-glutaryl-CoA reductase expression and increasing cholesterol 7α-hydroxylase expression. Moreover, FSDF also controls obesity development by lowering total cholesterol and low-density lipoprotein cholesterol levels in the blood, triglyceride, total cholesterol, and bile acid levels in the liver. Notably, FSDF supplements can promote Bacteroides and Prevotella propagation; excretive propionic acid binds to free fatty acid receptor 2/3 and then stimulates intestinal epithelial cells to generate glucagon-like-peptide-1 and peptide YY, which can reduce food and energy intake and ultimately prevent obesity. All evidence suggests that FSDF supplements play a crucial role in preventing obesity.
Collapse
Affiliation(s)
- Duo Yang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Juan Shen
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Chao Tang
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu Province, China
| | - Zhaoxin Lu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Fengxia Lu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Xiaomei Bie
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Fanqiang Meng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Haizhen Zhao
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China.
| |
Collapse
|
8
|
Yang S, Liu C, Jiang M, Liu X, Geng L, Zhang Y, Sun S, Wang K, Yin J, Ma S, Wang S, Belmonte JCI, Zhang W, Qu J, Liu GH. A single-nucleus transcriptomic atlas of primate liver aging uncovers the pro-senescence role of SREBP2 in hepatocytes. Protein Cell 2024; 15:98-120. [PMID: 37378670 PMCID: PMC10833472 DOI: 10.1093/procel/pwad039] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 05/19/2023] [Indexed: 06/29/2023] Open
Abstract
Aging increases the risk of liver diseases and systemic susceptibility to aging-related diseases. However, cell type-specific changes and the underlying mechanism of liver aging in higher vertebrates remain incompletely characterized. Here, we constructed the first single-nucleus transcriptomic landscape of primate liver aging, in which we resolved cell type-specific gene expression fluctuation in hepatocytes across three liver zonations and detected aberrant cell-cell interactions between hepatocytes and niche cells. Upon in-depth dissection of this rich dataset, we identified impaired lipid metabolism and upregulation of chronic inflammation-related genes prominently associated with declined liver functions during aging. In particular, hyperactivated sterol regulatory element-binding protein (SREBP) signaling was a hallmark of the aged liver, and consequently, forced activation of SREBP2 in human primary hepatocytes recapitulated in vivo aging phenotypes, manifesting as impaired detoxification and accelerated cellular senescence. This study expands our knowledge of primate liver aging and informs the development of diagnostics and therapeutic interventions for liver aging and associated diseases.
Collapse
Affiliation(s)
- Shanshan Yang
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Chengyu Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengmeng Jiang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Xiaoqian Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Lingling Geng
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yiyuan Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Shuhui Sun
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Kang Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jian Yin
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuai Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | | | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Aging Biomarker Consortium, Beijing 100101, China
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- Aging Biomarker Consortium, Beijing 100101, China
| | - Guang-Hui Liu
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Xuanwu Hospital Capital Medical University, Beijing 100053, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- Aging Biomarker Consortium, Beijing 100101, China
| |
Collapse
|
9
|
Guo Y, Sun Q, Wang S, Zhang M, Lei Y, Wu J, Wang X, Hu W, Meng H, Li Z, Xu L, Huang F, Qiu Z. Corydalis saxicola Bunting total alkaloids improve NAFLD by suppressing de novo lipogenesis through the AMPK-SREBP1 axis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117162. [PMID: 37690477 DOI: 10.1016/j.jep.2023.117162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/24/2023] [Accepted: 09/07/2023] [Indexed: 09/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Along with the gradually increasing incidence, nonalcoholic fatty liver disease (NAFLD) has already been influencing the health of more and more people in the world. Corydalis saxicola Bunting (CSB), a valuable folk medicine, is the dried whole grass of a perennial herb, Yanhuanglian (Papaveraceae), which has significant effects on various hepatitis, liver fibrosis, cirrhosis and other liver diseases. Corydalis saxicola Bunting total alkaloids (CSBTA), a mixture of alkaloids extracted from CSB, exhibit widely-accepted hepatoprotective effects. AIM OF THE STUDY This study aimed to explore the therapeutic potential of CSBTA on NAFLD and the underlying mechanism. MATERIALS AND METHODS A mice model was established by high fat and high cholesterol diet (HFHCD) to study the benefits of CSBTA on the progression of NAFLD. The efficacy of CSBTA on NAFLD was revealed systematically via RNA-sequencing analysis. Further efficacy and molecular mechanism study were explored in mouse primary hepatocytes and HepG2 cells stimulated with high energy with or without pharmacological inhibition or gene silencing. RESULTS CSBTA effectively improved the major hallmarks of NAFLD including liver lipid accumulation, liver injury, inflammation and fibrosis in HFHCD-fed mice. RNA sequencing and targeted qPCR analysis jointly evidenced CSBTA significantly suppressed the expression of Srebf1, Acc1 and Fasn which are the genes responsible for fatty acid biosynthesis. Moreover, stable isotope tracer test denoted CSBTA reduced lipid accumulation via interrupting fatty acid biosynthesis in hepatocytes or the liver. Mechanistically, CSBTA could impede SREBP1 maturation via AMPK activation, thereby reducing DNL-derived lipid accumulation in hepatocytes. CONCLUSIONS CSBTA protected against hepatic steatosis and other hallmarks of NAFLD induced by HFHCD via suppressing DNL, through modulating the AMPK-SREBP1 axis. CSBTA may therefore have a therapeutic potential for NAFLD treatment.
Collapse
Affiliation(s)
- Yating Guo
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Qiushuang Sun
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Shijiao Wang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Mengdi Zhang
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Yuanyuan Lei
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Jiejie Wu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Xinhong Wang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Wenjun Hu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Haitao Meng
- Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China.
| | - Zhiyu Li
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Luzhou Xu
- Gastroenterology Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| | - Fang Huang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Zhixia Qiu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
10
|
Wang X, Chen Y, Meng H, Meng F. SREBPs as the potential target for solving the polypharmacy dilemma. Front Physiol 2024; 14:1272540. [PMID: 38269061 PMCID: PMC10806128 DOI: 10.3389/fphys.2023.1272540] [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: 09/04/2023] [Accepted: 12/26/2023] [Indexed: 01/26/2024] Open
Abstract
The phenomenon of polypharmacy is a common occurrence among older people with multiple health conditions due to the rapid increase in population aging and the popularization of clinical guidelines. The prevalence of metabolic syndrome is growing quickly, representing a serious threat to both the public and the worldwide healthcare systems. In addition, it enhances the risk of cardiovascular disease as well as mortality and morbidity. Sterol regulatory element binding proteins (SREBPs) are basic helix-loop-helix leucine zipper transcription factors that transcriptionally modulate genes that regulate lipid biosynthesis and uptake, thereby serving an essential role in biological systems regulation. In this article, we have described the structure of SREBPs and explored their activation and regulation of signals. We also reveal that SREBPs are intricately involved in the modulation of metabolic diseases and thus have tremendous potential as the novel target for single-drug therapy for multiple diseases.
Collapse
Affiliation(s)
| | | | | | - Fanbo Meng
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis (Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute), Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
11
|
Chen X, Zhang P, Ma W, Pan H, Hong W, Chen G, Ding H, Tang W, Lin G, Zhang Z. Protective effects of methyl protodioscin against lipid disorders and liver injury in hyperlipidemic gerbils. Heliyon 2023; 9:e22785. [PMID: 38089978 PMCID: PMC10711193 DOI: 10.1016/j.heliyon.2023.e22785] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 11/18/2023] [Accepted: 11/20/2023] [Indexed: 06/13/2024] Open
Abstract
Methyl protodioscin (MPD) is the main component of total diosgenin, which was reported to reduce cholesterol and triglyceride levels potentially. This study aimed to investigate the beneficial effects of MPD against lipid disorder in hyperlipidemic gerbils induced by a high-fat diet (HFD). Hyperlipidemia was induced in gerbils by feeding them with HFD for six weeks, and a daily oral dose of MPD solution (25 and 50 mg/kg/day) was administered. This study investigated blood lipid levels and hepatic lipid accumulation in hyperlipidemic gerbils. The potential mechanism of MPD was explored by detecting the expression level of genes, including SREBPs, ACC, FASN, HMGCR, PCSK9, and LDL-R. The results showed that MPD treatment decreased the body weight, the relative weight of the liver, blood lipid, and hepatic lipid levels of gerbils fed with HFD. The administration of MPD alleviates liver steatosis and injury in gerbils fed with an HFD. MPD treatment reduced the expression of HMGCR, increased the expression of LDL-R, and decreased the expression of PCSK9 for cholesterol reduction. Additionally, MPD treatment reduced the expression of hepatic ACC and FASN for triglycerides reduction. The underlying mechanisms for these effects are attributed to MPD-induced inhibition of protein expression of LXR, SREBP1, and SREBP2. This study demonstrates that MPD protects gerbils against lipid disorders and liver injury by suppressing hepatic SREBPs expression.
Collapse
Affiliation(s)
- Xiaojia Chen
- Department of Human Anatomy, School of Basic Medicine, Guangdong Medical University, Dongguan, 523808, China
| | - Pengfei Zhang
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Weilie Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangdong Medical University, Dongguan, 523808, China
| | - Haiqiang Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangdong Medical University, Dongguan, 523808, China
| | - Weitao Hong
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangdong Medical University, Dongguan, 523808, China
| | - Gengji Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangdong Medical University, Dongguan, 523808, China
| | - Hang Ding
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangdong Medical University, Dongguan, 523808, China
| | - Wanze Tang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangdong Medical University, Dongguan, 523808, China
| | - Guorong Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangdong Medical University, Dongguan, 523808, China
| | - Zhizhen Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangdong Medical University, Dongguan, 523808, China
| |
Collapse
|
12
|
Dang R, Wang J, Tang M, Han W, Jiang P. Vitamin D Receptor Activation Attenuates Olanzapine-Induced Dyslipidemia in Mice Through Alleviating Hepatic Endoplasmic Reticulum Stress. Adv Biol (Weinh) 2023; 7:e2300228. [PMID: 37565702 DOI: 10.1002/adbi.202300228] [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: 06/19/2023] [Revised: 08/02/2023] [Indexed: 08/12/2023]
Abstract
The involvement of vitamin D (VD) signaling in atypical antipsychotics (AAPs)-induced metabolic disturbances has been previously established. This study aims to elucidate the role of VD in maintaining endoplasmic reticulum (ER) homeostasis and its impact on AAPs-induced metabolic adverse effects. Female C57BL/6 mice receive either calcitriol or vehicle one week prior to co-treatment with olanzapine (OLZ) for an additional four weeks. Metabolic parameters, hepatic ER homeostasis, and the SREBPs pathway are assessed through biochemical assays and protein expression profiling. HepG2 cells are transfected with vitamin D receptor (VDR) siRNA for VDR knockdown. OLZ-treated HepG2 cells are exposed to calcitriol to examine its effects on SREBPs and the unfolded protein response (UPR) pathways. VDR activation by calcitriol reduces OLZ-induced hepatic ER stress, leading to decreased SREBPs activity and lipid accumulation. Conversely, the knockdown of VDR in HepG2 cells diminishes the protective effects of calcitriol against OLZ-induced ER stress and SREBPs activation. This resulted in sustained UPR activation, elevated cleaved SREBPs levels, and increased lipid accumulation. These findings highlight an essential role of VDR signaling in the beneficial effects of VD on OLZ-induced metabolic side effects. Targeting VDR to resolve ER stress is likely an applicable therapeutic strategy for AAPs-induced metabolic disturbances.
Collapse
Affiliation(s)
- Ruili Dang
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China
| | - Jing Wang
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China
| | - Mimi Tang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Wenxiu Han
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China
| |
Collapse
|
13
|
Lu J, Chen S, Bai X, Liao M, Qiu Y, Zheng LL, Yu H. Targeting cholesterol metabolism in Cancer: From molecular mechanisms to therapeutic implications. Biochem Pharmacol 2023; 218:115907. [PMID: 37931664 DOI: 10.1016/j.bcp.2023.115907] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/08/2023]
Abstract
Cholesterol is an essential component of cell membranes and helps to maintain their structure and function. Abnormal cholesterol metabolism has been linked to the development and progression of tumors. Changes in cholesterol metabolism triggered by internal or external stimuli can promote tumor growth. During metastasis, tumor cells require large amounts of cholesterol to support their growth and colonization of new organs. Recent research has shown that cholesterol metabolism is reprogrammed during tumor development, and this can also affect the anti-tumor activity of immune cells in the surrounding environment. However, identifying the specific targets in cholesterol metabolism that regulate cancer progression and the tumor microenvironment is still a challenge. Additionally, exploring the potential of combining statin drugs with other therapies for different types of cancer could be a worthwhile avenue for future drug development. In this review, we focus on the molecular mechanisms of cholesterol and its derivatives in cell metabolism and the tumor microenvironment, and discuss specific targets and relevant therapeutic agents that inhibit aspects of cholesterol homeostasis.
Collapse
Affiliation(s)
- Jia Lu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Siwei Chen
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xuejiao Bai
- Department of Anesthesiology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Minru Liao
- Department of Anesthesiology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuling Qiu
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.
| | - Ling-Li Zheng
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China.
| | - Haiyang Yu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
14
|
Jeon YG, Kim YY, Lee G, Kim JB. Physiological and pathological roles of lipogenesis. Nat Metab 2023; 5:735-759. [PMID: 37142787 DOI: 10.1038/s42255-023-00786-y] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 03/15/2023] [Indexed: 05/06/2023]
Abstract
Lipids are essential metabolites, which function as energy sources, structural components and signalling mediators. Most cells are able to convert carbohydrates into fatty acids, which are often converted into neutral lipids for storage in the form of lipid droplets. Accumulating evidence suggests that lipogenesis plays a crucial role not only in metabolic tissues for systemic energy homoeostasis but also in immune and nervous systems for their proliferation, differentiation and even pathophysiological roles. Thus, excessive or insufficient lipogenesis is closely associated with aberrations in lipid homoeostasis, potentially leading to pathological consequences, such as dyslipidaemia, diabetes, fatty liver, autoimmune diseases, neurodegenerative diseases and cancers. For systemic energy homoeostasis, multiple enzymes involved in lipogenesis are tightly controlled by transcriptional and post-translational modifications. In this Review, we discuss recent findings regarding the regulatory mechanisms, physiological roles and pathological importance of lipogenesis in multiple tissues such as adipose tissue and the liver, as well as the immune and nervous systems. Furthermore, we briefly introduce the therapeutic implications of lipogenesis modulation.
Collapse
Affiliation(s)
- Yong Geun Jeon
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Ye Young Kim
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Gung Lee
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Jae Bum Kim
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea.
| |
Collapse
|
15
|
Du Y, Xi M, Li Y, Zheng R, Ding X, Li X, Zhang X, Wang L, Xing J, Hong B. Tilianin improves lipid profile and alleviates atherosclerosis in ApoE -/- mice through up-regulation of SREBP2-mediated LDLR expression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 109:154577. [PMID: 36610166 DOI: 10.1016/j.phymed.2022.154577] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 09/15/2022] [Accepted: 11/22/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND The huge global burden of atherosclerotic cardiovascular diseases (CVDs) represents an urgent unmet need for the development of novel therapeutics. Dracocephalum moldavica L. has been used as a traditional Uygur medicine to treat various CVDs for centuries. Tilianin is a major flavonoid component of D. moldavica L. and has potential for preventing atherosclerosis. However, the molecular mechanisms that tilianin attenuate atherosclerosis are far from fully understood. PURPOSES The purpose of this study is to investigate the efficiency and underlying mechanisms of tilianin in controlling lipid profile and preventing atherogenesis. METHODS The lipid-lowering effect of tilianin was evaluated in C57BL/6 and ApoE-/- mice by systematically determining serum biochemical parameters. The effects of tilianin on the atherosclerotic lesion were observed in aortic roots and whole aortas of ApoE-/- mice with oil red O staining. Caecal content from ApoE-/- mice were collected for 16S rRNA gene sequence analysis to assess the structure of the gut microbiota. The inhibition of hepatosteatosis was verified by histological examination, and a liver transcriptome analysis was performed to elucidate the tilianin-induced hepatic transcriptional alterations. Effects of tilianin on the expression and function of LDLR were examined in HepG2 cells and ApoE-/- mice. Further mechanisms underlying the efficacy of tilianin were investigated in HepG2 cells. RESULTS Tilianin treatment improved lipid profiles in C57BL/6 and dyslipidemic ApoE-/- mice, especially reducing the serum LDL-cholesterol (LDL-C) level. Significant reductions of atherosclerotic lesion area and hepatosteatosis were observed in tilianin-treated ApoE-/- mice. The altered gut microbial composition in tilianin groups was associated with lipid metabolism and atherosclerosis. The liver transcriptome revealed that tilianin regulated the transcription of lipid metabolism-related genes. Then both in vitro and in vivo analyses revealed the potent effect of tilianin to enhance hepatic LDLR expression and its mediated LDL-C uptake. Further studies confirmed a critical role of SREBP2 in hepatic LDLR up-regulation by tilianin via increasing precursor and thus mature nuclear SREBP2 level. CONCLUSION This study demonstrated the lipid-lowering effect of tilianin through SREBP2-mediated transcriptional activation of LDLR. Our findings reveal a novel anti-atherosclerotic mechanism of tilianin and underlie its potential clinical use in modulating CVDs with good availability and affordability.
Collapse
Affiliation(s)
- Yu Du
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; CAMS Key Laboratory of Synthetic Biology for Drug Innovation, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Mei Xi
- Xinjiang Key Laboratory of Uighur Medicines, Xinjiang Institute of Materia Medica, Urumchi 830004, China
| | - Yihua Li
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ruifang Zheng
- Xinjiang Key Laboratory of Uighur Medicines, Xinjiang Institute of Materia Medica, Urumchi 830004, China
| | - Xiaotian Ding
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xingxing Li
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; CAMS Key Laboratory of Synthetic Biology for Drug Innovation, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiumin Zhang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Li Wang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jianguo Xing
- Xinjiang Key Laboratory of Uighur Medicines, Xinjiang Institute of Materia Medica, Urumchi 830004, China.
| | - Bin Hong
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; CAMS Key Laboratory of Synthetic Biology for Drug Innovation, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
16
|
Marelli G, Morina N, Portale F, Pandini M, Iovino M, Di Conza G, Ho PC, Di Mitri D. Lipid-loaded macrophages as new therapeutic target in cancer. J Immunother Cancer 2022; 10:jitc-2022-004584. [PMID: 35798535 PMCID: PMC9263925 DOI: 10.1136/jitc-2022-004584] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2022] [Indexed: 11/04/2022] Open
Abstract
Macrophages are main players of the innate immune system. They show great heterogeneity and play diverse functions that include support to development, sustenance of tissue homeostasis and defense against infections. Dysfunctional macrophages have been described in multiple pathologies including cancer. Indeed tumor-associated macrophages (TAMs) are abundant in most tumors and sustain cancer growth, promote invasion and mediate immune evasion. Importantly, lipid metabolism influences macrophage activation and lipid accumulation confers pathogenic features on macrophages. Notably, a subset of lipid-loaded macrophages has been recently identified in many tumor types. Lipid-loaded TAMs support tumor growth and progression and exert immune-suppressive activities. In this review, we describe the role of lipid metabolism in macrophage activation in physiology and pathology and we discuss the impact of lipid accumulation in macrophages in the context of cancer.
Collapse
Affiliation(s)
- Giulia Marelli
- Tumor Microenvironment Unit, IRCCS Humanitas Research Hospital, Lombardia, Italy
| | - Nicolò Morina
- Tumor Microenvironment Unit, IRCCS Humanitas Research Hospital, Lombardia, Italy.,Department of Biomedical Sciences, Humanitas University, Lombardia, Italy
| | - Federica Portale
- Tumor Microenvironment Unit, IRCCS Humanitas Research Hospital, Lombardia, Italy
| | - Marta Pandini
- Tumor Microenvironment Unit, IRCCS Humanitas Research Hospital, Lombardia, Italy.,Department of Biomedical Sciences, Humanitas University, Lombardia, Italy
| | - Marta Iovino
- Tumor Microenvironment Unit, IRCCS Humanitas Research Hospital, Lombardia, Italy
| | - Giusy Di Conza
- Department of Oncology, University of Lausanne, Lausanne, Switzerland.,Ludwig Institute of Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Ping-Chih Ho
- Department of Oncology, University of Lausanne, Lausanne, Switzerland.,Ludwig Institute of Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Diletta Di Mitri
- Tumor Microenvironment Unit, IRCCS Humanitas Research Hospital, Lombardia, Italy .,Department of Biomedical Sciences, Humanitas University, Lombardia, Italy
| |
Collapse
|
17
|
Chen L, Guo Y, Wu Z, Zhao S, Zhang Z, Zheng F, Sun L, Hao Z, Xu C, Wang T, Peng Y. Epicatechin gallate prevents the de novo synthesis of fatty acid and the migration of prostate cancer cells. Acta Biochim Biophys Sin (Shanghai) 2021; 53:1662-1669. [PMID: 34718375 DOI: 10.1093/abbs/gmab144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Indexed: 12/25/2022] Open
Abstract
Lipid metabolism disorder caused by the upregulation of lipogenic genes is a typical feature of prostate cancer. The synthesis of fatty acids is enhanced to accelerate the development of prostate cancer and is considered as a potential therapeutic target. Epicatechin gallate, an active compound of green tea, has been reported to modulate lipid metabolism. In this research, the potential role of epicatechin gallate in prostate cancer cells was evaluated. The results indicated that epicatechin gallate downregulates the expression of acetyl-CoA carboxylase, ATP citrate lyase, and fatty acid synthase in prostate cancer cells and prostate xenograft tissues, suggesting that epicatechin gallate can inhibit de novo fatty acid synthesis. Moreover, epicatechin gallate significantly restrains the migration rather than the viability of prostate cancer cells. PI3K/AKT/mTOR signaling pathway, which exhibits regulatory effect on lipogenesis, is also inhibited under epicatechin gallate treatment, while pretreatment with AKT activator SC79 or mTOR activator MHY1485 blocks the inhibitory effect of epicatechin gallate on the expression of lipogenic genes and the migration of prostate cancer cells. In conclusion, this study revealed that epicatechin gallate impairs the synthesis of fatty acids via inhibition PI3K/AKT/mTOR signaling pathway and then attenuates the migration of prostate cancer cells.
Collapse
Affiliation(s)
- Luyao Chen
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yaping Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zixuan Wu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuwu Zhao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhaiyi Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Fang Zheng
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Likang Sun
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zheng Hao
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Chen Xu
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin 300121, China
| | - Tao Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yanfei Peng
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
18
|
Xu X, Sun S, Liang L, Lou C, He Q, Ran M, Zhang L, Zhang J, Yan C, Yuan H, Zhou L, Chen X, Dai X, Wang B, Zhang J, Zhao J. Role of the Aryl Hydrocarbon Receptor and Gut Microbiota-Derived Metabolites Indole-3-Acetic Acid in Sulforaphane Alleviates Hepatic Steatosis in Mice. Front Nutr 2021; 8:756565. [PMID: 34722615 PMCID: PMC8548612 DOI: 10.3389/fnut.2021.756565] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/17/2021] [Indexed: 12/15/2022] Open
Abstract
Scope: Gut microbiome-derived metabolites are the major mediators of diet-induced host-microbial interactions. Aryl hydrocarbon receptor (AHR) plays a crucial role in glucose, lipid, and cholesterol metabolism in the liver. In this study, we aimed to investigate the role of indole-3-acetic acid (IAA) and AHR in sulforaphane (SFN) alleviates hepatic steatosis in mice fed on a high-fat diet (HFD). Methods and Results: The HFD-fed male C57BL/6 mice were intervened with SFN for 6 weeks. HFD-mice showed classical pathophysiological characteristics of hepatic steatosis. The results showed that SFN significantly reduced body weight, liver inflammation and hepatic steatosis in HFD-fed mice. SFN reduced hepatic lipogenesis by activating AHR/SREBP-1C pathway, which was confirmed in HepG2 cell experiments. Moreover, SFN increased hepatic antioxidant activity by modulating Nrf-2/NQO1 expression. SFN increased serum and liver IAA level in HFD mice. Notably, SFN manipulated the gut microbiota, resulting in reducing Deferribacteres and proportions of the phylum Firmicutes/Bacteroidetes and increasing the abundance of specific bacteria that produce IAA. Furthermore, SFN upregulated Ahr expression and decreased the expression of inflammatory cytokines in Raw264.7 cells. Conclusions: SFN ameliorated hepatic steatosis not only by modulating lipid metabolism via AHR/SREBP-1C pathway but regulating IAA and gut microbiota in HFD-induced NAFLD mice.
Collapse
Affiliation(s)
- Xiuxiu Xu
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China.,NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Siyuan Sun
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Ling Liang
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Chenxi Lou
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Qijin He
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Maojuan Ran
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Lu Zhang
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingyue Zhang
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Chen Yan
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Hengjie Yuan
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Lu Zhou
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Chen
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Dai
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Bangmao Wang
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Jie Zhang
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingwen Zhao
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
19
|
Li L, Yang J, Li F, Gao F, Zhu L, Hao J. FBXW7 mediates high glucose‑induced SREBP‑1 expression in renal tubular cells of diabetic nephropathy under PI3K/Akt pathway regulation. Mol Med Rep 2021; 23:233. [PMID: 33537812 PMCID: PMC7893693 DOI: 10.3892/mmr.2021.11872] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022] Open
Abstract
Diabetic nephropathy (DN) is a severe complication of diabetes mellitus and lipid metabolism abnormality serves a key role in the pathogenesis of DN. Sterol regulatory element-binding protein 1 (SREBP-1) overexpression mediates aberrant lipid accumulation in renal tubular cells of DN. However, the exact mechanism involved in increased SREBP-1 has not been fully elucidated. The aim of the present study was to explore the mechanism involved in SREBP-1 upregulation. Diabetic mice and high glucose-cultured HKC cells were chosen to detect the expression of FBXW7 and SREBP-1 using immunohistochemistry, western blotting and PCR. The present study demonstrated that F-box and WD repeat domain containing 7 (FBXW7) expression was decreased in renal tubular cells of diabetic mice. Moreover, the co-expression of FBXW7 and SREBP-1 was observed in renal tubular cells, but not in the glomeruli. High glucose-induced the downregulation of FBXW7 expression in in vitro cultured HKC cells, which was accompanied by SREBP-1 upregulation. In addition, overexpression of FBXW7 in HKC cells led to SREBP-1 downregulation. By contrast, knockdown of FBXW7 caused SREBP-1 upregulation in HKC cells. It was found that the PI3K/Akt signaling pathway was activated in high glucose-stimulated HKC cells, and inhibition of PI3K/Akt pathway using LY294002 increased FBXW7 expression and decreased SREBP-1 expression. Taken together, the present results suggested that FBXW7 mediated high glucose-induced SREBP-1 expression in renal tubular cells of DN, under the regulation of the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Lisha Li
- Department of Pathology, Cangzhou Hospital of Integrated TCM‑WM, Cangzhou, Hebei 061001, P.R. China
| | - Juxiang Yang
- The Office of Basic Medical College, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Fan Li
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Fan Gao
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Lin Zhu
- Department of Electromyogram, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050001, P.R. China
| | - Jun Hao
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| |
Collapse
|
20
|
Lipid Regulatory Proteins as Potential Therapeutic Targets for Ovarian Cancer in Obese Women. Cancers (Basel) 2020; 12:cancers12113469. [PMID: 33233362 PMCID: PMC7700662 DOI: 10.3390/cancers12113469] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 02/07/2023] Open
Abstract
Obesity has become a recognized global epidemic that is associated with numerous comorbidities including type II diabetes, cardiovascular disease, hypertension, and cancer incidence and progression. Ovarian cancer (OvCa) has a unique mechanism of intra-peritoneal metastasis, already present in 80% of women at the time of diagnosis, making it the fifth leading cause of death from gynecological malignancy. Meta-analyses showed that obesity increases the risk of OvCa progression, leads to enhanced overall and organ-specific tumor burden, and adversely effects survival of women with OvCa. Recent data discovered that tumors grown in mice fed on a western diet (40% fat) have elevated lipid levels and a highly increased expression level of sterol regulatory element binding protein 1 (SREBP1). SREBP1 is a master transcription factor that regulates de novo lipogenesis and lipid homeostasis, and induces lipogenic reprogramming of tumor cells. Elevated SREBP1 levels are linked to cancer cell proliferation and metastasis. This review will summarize recent findings to provide a current understanding of lipid regulatory proteins in the ovarian tumor microenvironment with emphasis on SREBP1 expression in the obese host, the role of SREBP1 in cancer progression and metastasis, and potential therapeutic targeting of SREBPs and SREBP-pathway genes in treating cancers, particularly in the context of host obesity.
Collapse
|
21
|
Kumar S, Verma AK, Rani R, Sharma A, Wang J, Shah SA, Behari J, Gonzalez RS, Kohli R, Gandhi CR. Hepatic Deficiency of Augmenter of Liver Regeneration Predisposes to Nonalcoholic Steatohepatitis and Fibrosis. Hepatology 2020; 72:1586-1604. [PMID: 32031683 PMCID: PMC8025692 DOI: 10.1002/hep.31167] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 01/22/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS The augmenter of liver regeneration (ALR) protein is critical for lipid homeostasis and mitochondrial function. We investigated high-fat/high-carbohydrate (HF/HC) diet-induced nonalcoholic fatty liver disease (NAFLD) in wild-type (WT), hepatocyte-specific ALR-knockout (ALR-H-KO), and ALR-heterozygous (ALR-H-HET) mice. ALR was measured in serum of human nonalcoholic steatohepatitis (NASH) and NASH-induced cirrhosis (serum and liver). APPROACH AND RESULTS HF/HC feeding decreased ALR expression in all groups of mice. The otherwise normal ALR-H-HET mice gained more weight and steatosis than WT mice when challenged metabolically with the HF/HC diet; ALR-H-KO mice gained the least weight and had the least steatosis. These findings were consistent with correspondingly increased triglycerides and cholesterol and altered expression of carnitine palmitoyltransferase 1a, sterol regulatory element-binding protein, acetyl coenzyme A carboxylase, and fatty acid synthase. All HF/HC-fed mice developed insulin resistance, the magnitude being lower in ALR-H-KO mice. HF/HC-fed ALR-H-HET mice were more resistant to glucose challenge than WT or ALR-H-KO mice. The frequency of tumor necrosis factor alpha-producing, interleukin 6 (IL6)-producing, and IL17-producing cells was greater in ALR-H-KO than ALR-H-HET and lowest in WT mice. HF/HC feeding did not increase their number in ALR-H-KO mice, and the increase in ALR-H-HET was greater than that in WT mice except for IL17 cells. Cluster of differentiation 25-positive (CD25+ ) forkhead box P3-positive CD4+ regulatory T-cell frequency was lower in ALR-H-HET than WT mice and further reduced in ALR-H-KO mice; HF/HC reduced regulatory T-cell frequency only in WT mice. HF/HC-fed ALR-H-HET, but not WT, mice developed fibrosis; and ALR-H-KO mice progressed to cirrhosis. White adipose tissue of HF/HC-fed ALR-deficient mice developed strong inflammation, indicating bidirectional interactions with the liver. Hepatic and serum ALR levels were significantly reduced in patients with NASH-cirrhosis. Serum ALR was also significantly lower in patients with NASH. CONCLUSIONS Hepatic ALR deficiency may be a critical predisposing factor for aggressive NAFLD progression.
Collapse
Affiliation(s)
- Sudhir Kumar
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA,Cincinnati VA Medical Center, Cincinnati, Ohio, USA
| | - Alok K. Verma
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA,Cincinnati VA Medical Center, Cincinnati, Ohio, USA
| | - Richa Rani
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA,Cincinnati VA Medical Center, Cincinnati, Ohio, USA
| | - Akanksha Sharma
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA,Cincinnati VA Medical Center, Cincinnati, Ohio, USA
| | - Jiang Wang
- Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Shimul A. Shah
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jaideep Behari
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rosa Salazar Gonzalez
- Division of Gastroenterology, Hepatology & Nutrition, Children’s Hospital Los Angeles, California, USA
| | - Rohit Kohli
- Division of Gastroenterology, Hepatology & Nutrition, Children’s Hospital Los Angeles, California, USA
| | - Chandrashekhar R. Gandhi
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA,Cincinnati VA Medical Center, Cincinnati, Ohio, USA,Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
22
|
NASH, Fibrosis and Hepatocellular Carcinoma: Lipid Synthesis and Glutamine/Acetate Signaling. Int J Mol Sci 2020; 21:ijms21186799. [PMID: 32947972 PMCID: PMC7555727 DOI: 10.3390/ijms21186799] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 12/21/2022] Open
Abstract
Primary liver cancer is predicted to be the sixth most common cancer and the fourth leading cause of cancer mortality worldwide. Recent studies identified nonalcoholic fatty liver disease (NAFLD) as the underlying cause in 13-38.2% of patients with hepatocellular carcinoma unrelated to viral hepatitis and alcohol abuse. NAFLD progresses to nonalcoholic steatohepatitis (NASH), which increases the risk for the development of liver fibrosis, cirrhosis, and hepatocellular carcinoma. NAFLD is characterized by dysregulation of lipid metabolism. In addition, lipid metabolism is effected not only in NAFLD, but also in a broad range of chronic liver diseases and tumor development. Cancer cells manipulate a variety of metabolic pathways, including lipid metabolism, in order to build up their own cellular components. Identifying tumor dependencies on lipid metabolism would provide options for novel targeting strategies. This review article summarizes the research evidence on metabolic reprogramming and focuses on lipid metabolism in NAFLD, NASH, fibrosis, and cancer. As alternative routes of acetyl-CoA production for fatty acid synthesis, topics on glutamine and acetate metabolism are included. Further, studies on small compound inhibitors targeting lipid metabolism are discussed. Understanding reprogramming strategies in liver diseases, as well as the visualization of the metabolism reprogramming networks, could uncover novel therapeutic options.
Collapse
|
23
|
Singh H, Sharma AK, Gupta M, Singh AP, Kaur G. Tinospora cordifolia attenuates high fat diet-induced obesity and associated hepatic and renal dysfunctions in rats. PHARMANUTRITION 2020. [DOI: 10.1016/j.phanu.2020.100189] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
24
|
Hishikawa D, Yanagida K, Nagata K, Kanatani A, Iizuka Y, Hamano F, Yasuda M, Okamura T, Shindou H, Shimizu T. Hepatic Levels of DHA-Containing Phospholipids Instruct SREBP1-Mediated Synthesis and Systemic Delivery of Polyunsaturated Fatty Acids. iScience 2020; 23:101495. [PMID: 32891885 PMCID: PMC7481256 DOI: 10.1016/j.isci.2020.101495] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/25/2020] [Accepted: 08/19/2020] [Indexed: 12/25/2022] Open
Abstract
Polyunsaturated fatty acids (PUFAs), such as docosahexaenoic acid (DHA) and arachidonic acid (ARA), play fundamental roles in mammalian physiology. Although PUFA imbalance causes various disorders, mechanisms of the regulation of their systemic levels are poorly understood. Here, we report that hepatic DHA-containing phospholipids (DHA-PLs) determine the systemic levels of PUFAs through the SREBP1-mediated transcriptional program. We demonstrated that liver-specific deletion of Agpat3 leads to a decrease of DHA-PLs and a compensatory increase of ARA-PLs not only in the liver but also in other tissues including the brain. Together with recent findings that plasma lysophosphatidylcholine (lysoPC) is the major source of brain DHA, our results indicate that hepatic AGPAT3 contributes to brain DHA accumulation by supplying DHA-PLs as precursors of DHA-lysoPC. Furthermore, dietary fish oil-mediated suppression of hepatic PUFA biosynthetic program was blunted in liver-specific Agpat3 deletion. Our findings highlight the central role of hepatic DHA-PLs as the molecular rheostat for systemic homeostasis of PUFAs.
Collapse
Affiliation(s)
- Daisuke Hishikawa
- Department of Lipid Signaling, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan.
| | - Keisuke Yanagida
- Department of Lipid Signaling, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Katsuyuki Nagata
- Department of Lipid Signaling, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Ayumi Kanatani
- Department of Lipid Signaling, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Yoshiko Iizuka
- Department of Lipid Signaling, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Fumie Hamano
- Department of Lipid Signaling, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan; Life Science Core Faculty, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Megumi Yasuda
- Department of Lipid Signaling, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Tadashi Okamura
- Laboratory Animal Medicine, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan; Section of Animal Models, Department of Infectious Diseases, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Hideo Shindou
- Department of Lipid Signaling, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan; Department of Lipid Science, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takao Shimizu
- Department of Lipid Signaling, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo 162-8655, Japan; Department of Lipidomics, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
25
|
Sapir A. Not So Slim Anymore-Evidence for the Role of SUMO in the Regulation of Lipid Metabolism. Biomolecules 2020; 10:E1154. [PMID: 32781719 PMCID: PMC7466032 DOI: 10.3390/biom10081154] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 12/11/2022] Open
Abstract
One of the basic building blocks of all life forms are lipids-biomolecules that dissolve in nonpolar organic solvents but not in water. Lipids have numerous structural, metabolic, and regulative functions in health and disease; thus, complex networks of enzymes coordinate the different compositions and functions of lipids with the physiology of the organism. One type of control on the activity of those enzymes is the conjugation of the Small Ubiquitin-like Modifier (SUMO) that in recent years has been identified as a critical regulator of many biological processes. In this review, I summarize the current knowledge about the role of SUMO in the regulation of lipid metabolism. In particular, I discuss (i) the role of SUMO in lipid metabolism of fungi and invertebrates; (ii) the function of SUMO as a regulator of lipid metabolism in mammals with emphasis on the two most well-characterized cases of SUMO regulation of lipid homeostasis. These include the effect of SUMO on the activity of two groups of master regulators of lipid metabolism-the Sterol Regulatory Element Binding Protein (SERBP) proteins and the family of nuclear receptors-and (iii) the role of SUMO as a regulator of lipid metabolism in arteriosclerosis, nonalcoholic fatty liver, cholestasis, and other lipid-related human diseases.
Collapse
Affiliation(s)
- Amir Sapir
- Department of Biology and the Environment, Faculty of Natural Sciences, University of Haifa-Oranim, Tivon 36006, Israel
| |
Collapse
|
26
|
Guo C, Xue H, Guo T, Zhang W, Xuan WQ, Ren YT, Wang D, Chen YH, Meng YH, Gao HL, Zhao P. Recombinant human lactoferrin attenuates the progression of hepatosteatosis and hepatocellular death by regulating iron and lipid homeostasis in ob/ob mice. Food Funct 2020; 11:7183-7196. [PMID: 32756704 DOI: 10.1039/d0fo00910e] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Lactoferrin (Lf), an iron-binding glycoprotein, has been shown to possess antioxidant and anti-inflammatory properties and exert modulatory effects on lipid homeostasis and non-alcoholic fatty liver disease (NAFLD), but our understanding of its regulatory mechanisms is limited and inconsistent. We used leptin-deficient (ob/ob) mice as the rodent model of NAFLD, and administered recombinant human Lf (4 mg per kg body weight) or control vehicle by intraperitoneal injection to evaluate the hepatoprotective effects of Lf. After 40 days of treatment with Lf, insulin sensitivity and hepatic steatosis in ob/ob mice were significantly improved with the down-regulation of sterol regulatory element binding protein-2 (SREBP2), indicating an improvement in hepatic lipid metabolism and function. We further explored the mechanism, and found that Lf may increase the hepatocellular iron output by targeting the hepcidin-ferroportin (FPn) axis, and then maintains the liver oxidative balance through a nonenzymatic antioxidant system, ultimately suppressing the death of hepatocytes. In addition, the cytoprotective role of Lf may be associated with the inhibition of endoplasmic reticulum (ER) stress and inflammation, promotion of autophagy of damaged hepatocytes and induction of up-regulation of hypoxia inducible factor-1α/vascular endothelial growth factor (HIF-lα/VEGF) to facilitate liver function recovery. These findings suggest that recombinant human Lf might be a potential therapeutic agent for mitigating or delaying the pathological process of NAFLD.
Collapse
Affiliation(s)
- Chuang Guo
- College of Life and Health Sciences, Northeastern University, No. 195, Chuangxin Road, Hunnan District, Shenyang, 110169, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
The Hepatic Plasma Membrane Citrate Transporter NaCT (SLC13A5) as a Molecular Target for Metformin. Sci Rep 2020; 10:8536. [PMID: 32444674 PMCID: PMC7244496 DOI: 10.1038/s41598-020-65621-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 04/23/2020] [Indexed: 12/17/2022] Open
Abstract
Metformin is the first-line treatment for type 2 diabetes. Inhibition of hepatic gluconeogenesis is the primary contributor to its anti-diabetic effect. Metformin inhibits complex I and α-glycerophosphate shuttle, and the resultant increase in cytoplasmic NADH/NAD+ ratio diverts glucose precursors away from gluconeogenesis. These actions depend on metformin-mediated activation of AMP kinase (AMPK). Here we report on a hitherto unknown mechanism. Metformin inhibits the expression of the plasma membrane citrate transporter NaCT in HepG2 cells and decreases cellular levels of citrate. 5-Aminoimidazole-4-carboxamide ribonucleotide (AICAR), an AMPK activator, elicits a similar effect. The process involves a decrease in maximal velocity with no change in substrate affinity. The decrease in NaCT expression is associated with decreased mRNA levels. AMPK inhibits mTOR, and the mTOR inhibitor rapamycin also decreases NaCT expression. The transcription factor downstream of AMPK that is relevant to cAMP signaling is CREB; decreased levels of phospho-CREB seem to mediate the observed effects of metformin on NaCT. Citrate is known to suppress glycolysis by inhibiting phosphofructokinase-1 and activate gluconeogenesis by stimulating fructose-1,6-bisphophatase; therefore, the decrease in cellular levels of citrate would stimulate glycolysis and inhibit gluconeogenesis. These studies uncover a novel mechanism for the anti-diabetic actions of metformin.
Collapse
|
28
|
Zeng SL, Li SZ, Xiao PT, Cai YY, Chu C, Chen BZ, Li P, Li J, Liu EH. Citrus polymethoxyflavones attenuate metabolic syndrome by regulating gut microbiome and amino acid metabolism. SCIENCE ADVANCES 2020; 6:eaax6208. [PMID: 31922003 PMCID: PMC6941918 DOI: 10.1126/sciadv.aax6208] [Citation(s) in RCA: 266] [Impact Index Per Article: 53.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 11/07/2019] [Indexed: 05/19/2023]
Abstract
Metabolic syndrome (MetS) is intricately linked to dysregulation of gut microbiota and host metabolomes. Here, we first find that a purified citrus polymethoxyflavone-rich extract (PMFE) potently ameliorates high-fat diet (HFD)-induced MetS, alleviates gut dysbiosis, and regulates branched-chain amino acid (BCAA) metabolism using 16S rDNA amplicon sequencing and metabolomic profiling. The metabolic protective effects of PMFE are gut microbiota dependent, as demonstrated by antibiotic treatment and fecal microbiome transplantation (FMT). The modulation of gut microbiota altered BCAA levels in the host serum and feces, which were significantly associated with metabolic features and actively responsive to therapeutic interventions with PMFE. Notably, PMFE greatly enriched the commensal bacterium Bacteroides ovatus, and gavage with B. ovatus reduced BCAA concentrations and alleviated MetS in HFD mice. PMFE may be used as a prebiotic agent to attenuate MetS, and target-specific microbial species may have unique therapeutic promise for metabolic diseases.
Collapse
Affiliation(s)
- Su-Ling Zeng
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, China
| | - Shang-Zhen Li
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, China
| | - Ping-Ting Xiao
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, China
| | - Yuan-Yuan Cai
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, China
| | - Chu Chu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Bai-Zhong Chen
- Guangdong Xinbaotang Biological Technology Co. Ltd., Guangdong, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, China
- Corresponding author. (E.-H.L.); (J.L.); (P.L.)
| | - Jing Li
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, China
- Corresponding author. (E.-H.L.); (J.L.); (P.L.)
| | - E-Hu Liu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, China
- Corresponding author. (E.-H.L.); (J.L.); (P.L.)
| |
Collapse
|
29
|
Zhang Y, Li C, Hu C, Wu Q, Cai Y, Xing S, Lu H, Wang L, Huang D, Sun L, Li T, He X, Zhong X, Wang J, Gao P, Smith ZJ, Jia W, Zhang H. Lin28 enhances de novo fatty acid synthesis to promote cancer progression via SREBP-1. EMBO Rep 2019; 20:e48115. [PMID: 31379107 PMCID: PMC6776893 DOI: 10.15252/embr.201948115] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/03/2019] [Accepted: 07/17/2019] [Indexed: 12/15/2022] Open
Abstract
Lin28 plays an important role in promoting tumor development, whereas its exact functions and underlying mechanisms are largely unknown. Here, we show that both human homologs of Lin28 accelerate de novo fatty acid synthesis and promote the conversion from saturated to unsaturated fatty acids via the regulation of SREBP-1. By directly binding to the mRNAs of both SREBP-1 and SCAP, Lin28A/B enhance the translation and maturation of SREBP-1, and protect cancer cells from lipotoxicity. Lin28A/B-stimulated tumor growth is abrogated by SREBP-1 inhibition and by the impairment of the RNA binding properties of Lin28A/B, respectively. Collectively, our findings uncover that post-transcriptional regulation by Lin28A/B enhances de novo fatty acid synthesis and metabolic conversion of saturated and unsaturated fatty acids via SREBP-1, which is critical for cancer progression.
Collapse
Affiliation(s)
- Yang Zhang
- Anhui Key Laboratory of Hepatopancreatobiliary SurgeryDepartment of General SurgeryAnhui Provincial HospitalThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
- Hefei National Laboratory for Physical Sciences at MicroscaleThe CAS Key Laboratory of Innate Immunity and Chronic DiseaseDivision of Molecular MedicineSchool of Life Sciences and Medical CenterUniversity of Science and Technology of ChinaHefeiChina
| | - Chenchen Li
- Hefei National Laboratory for Physical Sciences at MicroscaleThe CAS Key Laboratory of Innate Immunity and Chronic DiseaseDivision of Molecular MedicineSchool of Life Sciences and Medical CenterUniversity of Science and Technology of ChinaHefeiChina
| | - Chuanzhen Hu
- Department of Precision Machinery and Precision InstrumentationUniversity of Science and Technology of ChinaHefeiChina
| | - Qian Wu
- Shanghai Center for Bioinformation TechnologyShanghaiChina
| | - Yongping Cai
- Department of PathologySchool of MedicineAnhui Medical UniversityHefeiChina
| | - Songge Xing
- Anhui Key Laboratory of Hepatopancreatobiliary SurgeryDepartment of General SurgeryAnhui Provincial HospitalThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
- Hefei National Laboratory for Physical Sciences at MicroscaleThe CAS Key Laboratory of Innate Immunity and Chronic DiseaseDivision of Molecular MedicineSchool of Life Sciences and Medical CenterUniversity of Science and Technology of ChinaHefeiChina
| | - Hui Lu
- Hefei National Laboratory for Physical Sciences at MicroscaleThe CAS Key Laboratory of Innate Immunity and Chronic DiseaseDivision of Molecular MedicineSchool of Life Sciences and Medical CenterUniversity of Science and Technology of ChinaHefeiChina
| | - Lin Wang
- Hefei National Laboratory for Physical Sciences at MicroscaleThe CAS Key Laboratory of Innate Immunity and Chronic DiseaseDivision of Molecular MedicineSchool of Life Sciences and Medical CenterUniversity of Science and Technology of ChinaHefeiChina
| | - De Huang
- Hefei National Laboratory for Physical Sciences at MicroscaleThe CAS Key Laboratory of Innate Immunity and Chronic DiseaseDivision of Molecular MedicineSchool of Life Sciences and Medical CenterUniversity of Science and Technology of ChinaHefeiChina
| | - Linchong Sun
- Laboratory of Cancer and Stem Cell MetabolismSchool of MedicineInstitutes for Life SciencesSouth China University of TechnologyGuangzhouChina
| | - Tingting Li
- Hefei National Laboratory for Physical Sciences at MicroscaleThe CAS Key Laboratory of Innate Immunity and Chronic DiseaseDivision of Molecular MedicineSchool of Life Sciences and Medical CenterUniversity of Science and Technology of ChinaHefeiChina
| | - Xiaoping He
- Hefei National Laboratory for Physical Sciences at MicroscaleThe CAS Key Laboratory of Innate Immunity and Chronic DiseaseDivision of Molecular MedicineSchool of Life Sciences and Medical CenterUniversity of Science and Technology of ChinaHefeiChina
| | - Xiuying Zhong
- Laboratory of Cancer and Stem Cell MetabolismSchool of MedicineInstitutes for Life SciencesSouth China University of TechnologyGuangzhouChina
| | - Junfeng Wang
- High Magnetic Field LaboratoryChinese Academy of SciencesHefeiChina
| | - Ping Gao
- Anhui Key Laboratory of Hepatopancreatobiliary SurgeryDepartment of General SurgeryAnhui Provincial HospitalThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
- Laboratory of Cancer and Stem Cell MetabolismSchool of MedicineInstitutes for Life SciencesSouth China University of TechnologyGuangzhouChina
| | - Zachary J Smith
- Department of Precision Machinery and Precision InstrumentationUniversity of Science and Technology of ChinaHefeiChina
| | - Weidong Jia
- Anhui Key Laboratory of Hepatopancreatobiliary SurgeryDepartment of General SurgeryAnhui Provincial HospitalThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
| | - Huafeng Zhang
- Anhui Key Laboratory of Hepatopancreatobiliary SurgeryDepartment of General SurgeryAnhui Provincial HospitalThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
- Hefei National Laboratory for Physical Sciences at MicroscaleThe CAS Key Laboratory of Innate Immunity and Chronic DiseaseDivision of Molecular MedicineSchool of Life Sciences and Medical CenterUniversity of Science and Technology of ChinaHefeiChina
| |
Collapse
|
30
|
Kusnadi A, Park SH, Yuan R, Pannellini T, Giannopoulou E, Oliver D, Lu T, Park-Min KH, Ivashkiv LB. The Cytokine TNF Promotes Transcription Factor SREBP Activity and Binding to Inflammatory Genes to Activate Macrophages and Limit Tissue Repair. Immunity 2019; 51:241-257.e9. [PMID: 31303399 PMCID: PMC6709581 DOI: 10.1016/j.immuni.2019.06.005] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 04/23/2019] [Accepted: 06/07/2019] [Indexed: 12/20/2022]
Abstract
Cytokine tumor necrosis factor (TNF)-mediated macrophage polarization is important for inflammatory disease pathogenesis, but the mechanisms regulating polarization are not clear. We performed transcriptomic and epigenomic analysis of the TNF response in primary human macrophages and revealed late-phase activation of SREBP2, the master regulator of cholesterol biosynthesis genes. TNF stimulation extended the genomic profile of SREBP2 occupancy to include binding to and activation of inflammatory and interferon response genes independently of its functions in sterol metabolism. Genetic ablation of SREBP function shifted the balance of macrophage polarization from an inflammatory to a reparative phenotype in peritonitis and skin wound healing models. Genetic ablation of SREBP activity in myeloid cells or topical pharmacological inhibition of SREBP improved skin wound healing under homeostatic and chronic inflammatory conditions. Our results identify a function and mechanism of action for SREBPs in augmenting TNF-induced macrophage activation and inflammation and open therapeutic avenues for promoting wound repair.
Collapse
Affiliation(s)
- Anthony Kusnadi
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA; Research Institute and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
| | - Sung Ho Park
- Research Institute and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA; School of Life Sciences, Ulsan National Institute of Science & Technology (UNIST), Ulsan 44919, Korea
| | - Ruoxi Yuan
- Research Institute and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
| | - Tania Pannellini
- Research Institute and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
| | - Eugenia Giannopoulou
- Research Institute and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA; Biological Sciences Department, New York City College of Technology, City University of New York, Brooklyn, NY 11201, USA
| | - David Oliver
- Research Institute and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
| | - Theresa Lu
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA; Research Institute and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
| | - Kyung-Hyun Park-Min
- BCMB Allied Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA; Research Institute and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA.
| | - Lionel B Ivashkiv
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA; Research Institute and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA.
| |
Collapse
|
31
|
Chrysin ameliorates nonalcoholic fatty liver disease in rats. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:1617-1628. [PMID: 31372694 DOI: 10.1007/s00210-019-01705-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 07/23/2019] [Indexed: 12/14/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is regarded as the hepatic manifestation of the metabolic syndrome. It begins with the accumulation of fat in the liver (simple steatosis), which if untreated can progress to nonalcoholic steatohepatitis, cirrhosis, and hepatocellular carcinoma. Chrysin is a flavonoid present in bee propolis and many other plants. The objective of this study was to determine if chrysin can ameliorate NAFLD induced by feeding a high fructose diet (HFD) in rats. The rats were divided into five groups: normal control, HFD control, chrysin (25, 50, and 100 mg/kg p.o. body weight). Biochemical estimations were carried out in the serum and liver of rats. The gene expressions of SREBP-1c and PPAR α were determined in the liver. The histopathology of the liver was also studied. Chrysin caused a significant decrease in the serum fasting glucose and improved the insulin resistance, dyslipidemia, and liver enzymes. It caused a significant decrease in the liver weight and hepatic free fatty acids, triglyceride, and cholesterol content. Chrysin exerted antioxidant effects, reduced carbonyl content, advanced glycation end products, collagen, TNF-α, and IL-6 concentrations in the liver. Chrysin significantly reduced the hepatic gene expression of SREBP-1c and increased that of PPAR-α. The histopathology of liver of rats treated with chrysin showed significant decrease in the steatosis, ballooning, and lobular inflammation when compared to the HFD control group. Thus, chrysin demonstrated anti-steatotic, antiglycating, antioxidant, anti-inflammatory, and antifibrotic effects and seems to be a promising molecule for the management of NAFLD.
Collapse
|
32
|
Li S, Yu W, Guan X, Huang K, Liu J, Liu D, Duan R. Effects of millet whole grain supplementation on the lipid profile and gut bacteria in rats fed with high-fat diet. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.05.030] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
33
|
Yang N, Li C, Li H, Liu M, Cai X, Cao F, Feng Y, Li M, Wang X. Emodin Induced SREBP1-Dependent and SREBP1-Independent Apoptosis in Hepatocellular Carcinoma Cells. Front Pharmacol 2019; 10:709. [PMID: 31297058 PMCID: PMC6607744 DOI: 10.3389/fphar.2019.00709] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 05/31/2019] [Indexed: 12/24/2022] Open
Abstract
Reynoutria multiflora (Thunb.) Moldenke (He Shou Wu) has been used for about 20 centuries as a Chinese medicinal herb for its activities of anticancer, anti-hyperlipidemia, and anti-aging. Previously, we found that He Shou Wu ethanol extract could induce apoptosis in hepatocellular carcinoma cells, and we also screened its active components. In this study, we investigated whether lowering lipid metabolism of emodin, a main active component in He Shou Wu, was associated with inhibitory effects in hepatocellular carcinoma cells. The correlation of apoptosis induction and lipid metabolism was investigated. The intrinsic apoptotic cell death, lipid production, and their signaling pathways were investigated in emodin-treated human hepatocellular carcinoma cells Bel-7402. The data showed that emodin triggered apoptosis in Bel-7402 cells. The mitochondrial membrane potential (ΔΨm) was reduced in emodin-treated Bel-7402 cells. We also found that emodin activated the expression of intrinsic apoptosis signaling pathway-related proteins, cleaved-caspase 9 and 3, Apaf 1, cytochrome c (CYTC), apoptosis-inducing factor, endonuclease G, Bax, and Bcl-2. Furthermore, the level of triglycerides and desaturation of fatty acids was reduced in Bel-7402 cells when exposed to emodin. Furthermore, the expression level of messenger RNA (mRNA) and protein of sterol regulatory element binding protein 1 (SREBP1) as well as its downstream signaling pathway and the synthesis and the desaturation of fatty acid metabolism-associated proteins (adenosine triphosphate citrate lyase, acetyl-CoA carboxylase alpha, fatty acid synthase (FASN), and stearoyl-CoA desaturase D) were also decreased. Notably, knock-out of SREBP1 in Bel-7402 cells was also found to induce less intrinsic apoptosis than did emodin. In conclusion, these results indicated that emodin could induce apoptosis in an SREBP1-dependent and SREBP1-independent manner in hepatocellular carcinoma cells.
Collapse
Affiliation(s)
- Nian Yang
- Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Biomedical Research Institute, Hubei University of Medicine, Shiyan, China.,Department of Pharmacy, Jurong Hospital Affiliated to Jiangsu University, Zhenjiang, China
| | - Chen Li
- Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Biomedical Research Institute, Hubei University of Medicine, Shiyan, China
| | - Hongliang Li
- Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Biomedical Research Institute, Hubei University of Medicine, Shiyan, China
| | - Ming Liu
- Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Biomedical Research Institute, Hubei University of Medicine, Shiyan, China
| | - Xiaojun Cai
- Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Fengjun Cao
- Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Yibin Feng
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
| | - Minglun Li
- Department of Radiation Oncology, University Hospital, LMU, Munich, Germany
| | - Xuanbin Wang
- Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Biomedical Research Institute, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
34
|
Lipid Metabolism as a Source of Druggable Targets for Antiviral Discovery against Zika and Other Flaviviruses. Pharmaceuticals (Basel) 2019; 12:ph12020097. [PMID: 31234348 PMCID: PMC6631711 DOI: 10.3390/ph12020097] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 06/14/2019] [Accepted: 06/19/2019] [Indexed: 02/07/2023] Open
Abstract
The Zika virus (ZIKV) is a mosquito-borne flavivirus that can lead to birth defects (microcephaly), ocular lesions and neurological disorders (Guillain-Barré syndrome). There is no licensed vaccine or antiviral treatment against ZIKV infection. The effort to understand the complex interactions of ZIKV with cellular networks contributes to the identification of novel host-directed antiviral (HDA) candidates. Among the cellular pathways involved in infection, lipid metabolism gains attention. In ZIKV-infected cells lipid metabolism attributed to intracellular membrane remodeling, virion morphogenesis, autophagy modulation, innate immunity and inflammation. The key roles played by the cellular structures associated with lipid metabolism, such as peroxisomes and lipid droplets, are starting to be deciphered. Consequently, there is a wide variety of lipid-related antiviral strategies that are currently under consideration, which include an inhibition of sterol regulatory element-binding proteins (SREBP), the activation of adenosine-monophosphate activated kinase (AMPK), an inhibition of acetyl-Coenzyme A carboxylase (ACC), interference with sphingolipid metabolism, blockage of intracellular cholesterol trafficking, or a treatment with cholesterol derivatives. Remarkably, most of the HDAs identified in these studies are also effective against flaviviruses other than ZIKV (West Nile virus and dengue virus), supporting their broad-spectrum effect. Considering that lipid metabolism is one of the main cellular pathways suitable for pharmacological intervention, the idea of repositioning drugs targeting lipid metabolism as antiviral candidates is gaining force.
Collapse
|
35
|
Matsuoka S, Bariuan JV, Nakagiri S, Abd Eldaim MA, Okamatsu-Ogura Y, Kimura K. Linking pathways and processes: Retinoic acid and glucose. MOLECULAR NUTRITION: CARBOHYDRATES 2019:247-264. [DOI: 10.1016/b978-0-12-849886-6.00013-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
36
|
Guo WL, Pan YY, Li L, Li TT, Liu B, Lv XC. Ethanol extract of Ganoderma lucidum ameliorates lipid metabolic disorders and modulates the gut microbiota composition in high-fat diet fed rats. Food Funct 2018; 9:3419-3431. [PMID: 29877551 DOI: 10.1039/c8fo00836a] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The objective of this study was to investigate the effects of ethanol extract of Ganoderma lucidum (GL95) on hyperlipidaemia and gut microbiota, and its regulation mechanism in Wistar rats fed on a high-fat diet (HFD). UPLC-QTOF MS indicated that GL95 was enriched with triterpenoids, especially ganoderic acids. The results of the animal experiment showed that oral administration of GL95 markedly alleviated the dyslipidemia through decreasing the levels of serum total triglyceride (TG), total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C), and inhibiting hepatic lipid accumulation and steatosis. Furthermore, GL95 supplementation altered the composition of gut microbiota, in particular modulating the relative abundance of functionally relevant enterotypes compared with the HFD group. The Spearman's correlation analysis revealed that Alistipes, Defluviitalea, Peptococcaceae and Alloprevotella were negatively correlated with serum and hepatic lipid profiles. Meanwhile, the GL95 treatment regulated the mRNA expression levels of the genes involved in lipid and cholesterol metabolism. The findings above illustrate that Ganoderma triterpenoids have the potential to ameliorate lipid metabolic disorders, in part through modulating specific gut microbiota and regulating the genes involved in lipid and cholesterol metabolism, suggesting Ganoderma triterpenoids as a potential novel functional food for the treatment or prevention of hyperlipidaemia.
Collapse
Affiliation(s)
- Wei-Ling Guo
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, P. R. China.
| | | | | | | | | | | |
Collapse
|
37
|
Moslehi A, Hamidi-zad Z. Role of SREBPs in Liver Diseases: A Mini-review. J Clin Transl Hepatol 2018; 6:332-338. [PMID: 30271747 PMCID: PMC6160306 DOI: 10.14218/jcth.2017.00061] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 01/10/2018] [Accepted: 02/11/2018] [Indexed: 12/15/2022] Open
Abstract
Sterol regulator element binding proteins (SREBPs) are a family of transcription factors involved in the biogenesis of cholesterol, fatty acids and triglycerides. They also regulate physiological functions of many organs, such as thyroid, brain, heart, pancreas and hormone synthesis. Beside the physiological effects, SREBPs participate in some pathological processes, diabetes, endoplasmic reticulum stress, atherosclerosis and chronic kidney disease associated with SREBP expression changes. In the liver, SREBPs are involved in the pathogenesis of nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, hepatitis and hepatic cancer. There are several SREBP inhibitors that have potential for treating obesity, diabetes and cancer. This review assesses the recent findings about the roles of SREBPs in the physiology of organs' function and pathogenesis of liver diseases.
Collapse
Affiliation(s)
- Azam Moslehi
- Department of Physiology, Cellular & Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Zeinab Hamidi-zad
- Department of Physiology, Cellular & Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| |
Collapse
|
38
|
Westfall S, Lomis N, Prakash S. A polyphenol-rich prebiotic in combination with a novel probiotic formulation alleviates markers of obesity and diabetes in Drosophila. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.07.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
39
|
Cheng X, Li J, Guo D. SCAP/SREBPs are Central Players in Lipid Metabolism and Novel Metabolic Targets in Cancer Therapy. Curr Top Med Chem 2018; 18:484-493. [PMID: 29788888 DOI: 10.2174/1568026618666180523104541] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/02/2017] [Accepted: 01/03/2018] [Indexed: 01/09/2023]
Abstract
Lipid metabolism reprogramming emerges as a new hallmark of malignancies. Sterol regulatory element-binding proteins (SREBPs), which are central players in lipid metabolism, are endoplasmic reticulum (ER)-bound transcription factors that control the expression of genes important for lipid synthesis and uptake. Their transcriptional activation requires binding to SREBP cleavageactivating protein (SCAP) to translocate their inactive precursors from the ER to the Golgi to undergo cleavage and subsequent nucleus translocation of their NH2-terminal forms. Recent studies have revealed that SREBPs are markedly upregulated in human cancers, providing the mechanistic link between lipid metabolism alterations and malignancies. Pharmacological or genetic inhibition of SCAP or SREBPs significantly suppresses tumor growth in various cancer models, demonstrating that SCAP/SREBPs could serve as promising metabolic targets for cancer therapy. In this review, we will summarize recent progress in our understanding of the underlying molecular mechanisms regulating SCAP/SREBPs and lipid metabolism in malignancies, discuss new findings about SREBP trafficking, which requires SCAP N-glycosylation, and introduce a newly identified microRNA-29-mediated negative feedback regulation of the SCAP/SREBP pathway. Moreover, we will review recently developed inhibitors targeting the SCAP/SREBP pathway for cancer treatment.
Collapse
Affiliation(s)
- Xiang Cheng
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Columbus, OH 43210, United States
| | - Jianying Li
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Columbus, OH 43210, United States
| | - Deliang Guo
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Columbus, OH 43210, United States
| |
Collapse
|
40
|
Sterol regulatory element binding protein 1 trans-activates 25-hydroxy vitamin D3 24-hydroxylase gene expression in renal proximal tubular cells. Biochem Biophys Res Commun 2018; 500:275-282. [DOI: 10.1016/j.bbrc.2018.04.058] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 04/09/2018] [Indexed: 01/25/2023]
|
41
|
Zhu J, Jin J, Ding J, Li S, Cen P, Wang K, Wang H, Xia J. Ganoderic Acid A improves high fat diet-induced obesity, lipid accumulation and insulin sensitivity through regulating SREBP pathway. Chem Biol Interact 2018; 290:77-87. [PMID: 29852127 DOI: 10.1016/j.cbi.2018.05.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 05/15/2018] [Accepted: 05/28/2018] [Indexed: 12/12/2022]
Abstract
Obesity and its major co-morbidity, type 2 diabetes, have been an alarming epidemic prevalence without an effective treatment available. Sterol regulatory element-binding proteins (SREBPs) are major transcription factors regulating the expression of genes involved in biosynthesis of cholesterol, fatty acid and triglyceride. Therefore, inhibition of SREBP pathway may be a useful strategy to treat obesity with type 2 diabetes. Here, we identify a small molecule, Ganoderic Acid A (GAA), inhibits the SREBP expression and decreases the cellular levels of cholesterol and fatty acid in vitro. GAA also ameliorates body weight gain and fat accumulation in liver or adipose tissues, and improves serum lipid levels and insulin sensitivity in high fat diet (HFD)-induced obese mice. Consistently, GAA regulates SREBPs target genes and metabolism associated genes in liver or adipose tissues, which may directly contribute to the lower lipid level and improvement of insulin resistance. Taken together, GAA could be a potential leading compound for development of drugs for the prevention of obesity and insulin resistance.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Infectious Diseases, Hangzhou First People's Hospital, Nanjing Medical University, 310006, Hangzhou, Zhejiang, China
| | - Jie Jin
- Department of Infectious Diseases, Hangzhou First People's Hospital, Nanjing Medical University, 310006, Hangzhou, Zhejiang, China
| | - Jiexia Ding
- Department of Infectious Diseases, Hangzhou First People's Hospital, Nanjing Medical University, 310006, Hangzhou, Zhejiang, China
| | - Siying Li
- Department of Infectious Diseases, Hangzhou First People's Hospital, Nanjing Medical University, 310006, Hangzhou, Zhejiang, China
| | - Panpan Cen
- Department of Infectious Diseases, Hangzhou First People's Hospital, Nanjing Medical University, 310006, Hangzhou, Zhejiang, China
| | - Keyi Wang
- Central Laboratory, Hangzhou First People's Hospital, Nanjing Medical University, 310006, Hangzhou, Zhejiang, China
| | - Hai Wang
- Department of Laboratory, Tongde Hospital of Zhejiang Province, 310012, Hangzhou, Zhejiang, China
| | - Junbo Xia
- Department of Pulmonary Medicine Hangzhou First People's Hospital, Nanjing Medical University, 310006, Hangzhou, Zhejiang, China.
| |
Collapse
|
42
|
Cheng C, Geng F, Cheng X, Guo D. Lipid metabolism reprogramming and its potential targets in cancer. Cancer Commun (Lond) 2018; 38:27. [PMID: 29784041 PMCID: PMC5993136 DOI: 10.1186/s40880-018-0301-4] [Citation(s) in RCA: 520] [Impact Index Per Article: 74.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/12/2018] [Indexed: 12/13/2022] Open
Abstract
Reprogramming of lipid metabolism is a newly recognized hallmark of malignancy. Increased lipid uptake, storage and lipogenesis occur in a variety of cancers and contribute to rapid tumor growth. Lipids constitute the basic structure of membranes and also function as signaling molecules and energy sources. Sterol regulatory element-binding proteins (SREBPs), a family of membrane-bound transcription factors in the endoplasmic reticulum, play a central role in the regulation of lipid metabolism. Recent studies have revealed that SREBPs are highly up-regulated in various cancers and promote tumor growth. SREBP cleavage-activating protein is a key transporter in the trafficking and activation of SREBPs as well as a critical glucose sensor, thus linking glucose metabolism and de novo lipid synthesis. Targeting altered lipid metabolic pathways has become a promising anti-cancer strategy. This review summarizes recent progress in our understanding of lipid metabolism regulation in malignancy, and highlights potential molecular targets and their inhibitors for cancer treatment.
Collapse
Affiliation(s)
- Chunming Cheng
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Columbus, OH, 43210, USA
| | - Feng Geng
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Columbus, OH, 43210, USA
| | - Xiang Cheng
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Columbus, OH, 43210, USA
| | - Deliang Guo
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Columbus, OH, 43210, USA.
| |
Collapse
|
43
|
Physiological functions of FBW7 in cancer and metabolism. Cell Signal 2018; 46:15-22. [PMID: 29474981 DOI: 10.1016/j.cellsig.2018.02.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 02/16/2018] [Accepted: 02/18/2018] [Indexed: 12/11/2022]
Abstract
FBW7 is one of the most well characterized F-box proteins that serve as substrate recognition subunits of SCF (Skp1-Cullin 1-F-box proteins) E3 ubiquitin ligase complexes. SCFFBW7 plays key roles in regulating cell cycle progression, differentiation, and stem cell maintenance largely through targeting a broad range of oncogenic substrates for proteasome-dependent degradation. The identification of an increasing number of FBW7 substrates for ubiquitination, and intensive in vitro and in vivo studies have revealed a network of signaling components controlled by FBW7 that contributes to metabolic regulation as well as its tumor suppressor role. Here we mainly focus on recent findings that highlight a critical role for FBW7 in cancer and metabolism.
Collapse
|
44
|
Zheng ZG, Lu C, Thu PM, Zhang X, Li HJ, Li P, Xu X. Praeruptorin B improves diet-induced hyperlipidemia and alleviates insulin resistance via regulating SREBP signaling pathway. RSC Adv 2018. [DOI: 10.1039/c7ra11797c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Many metabolic diseases are caused by disruption of lipid homeostasis.
Collapse
Affiliation(s)
- Zu-Guo Zheng
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing
- China
| | - Chong Lu
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing
- China
| | - Pyone Myat Thu
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing
- China
| | - Xin Zhang
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing
- China
| | - Hui-Jun Li
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing
- China
| | - Ping Li
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing
- China
| | - Xiaojun Xu
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing
- China
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
| |
Collapse
|
45
|
RNF20 Suppresses Tumorigenesis by Inhibiting the SREBP1c-PTTG1 Axis in Kidney Cancer. Mol Cell Biol 2017; 37:MCB.00265-17. [PMID: 28827316 DOI: 10.1128/mcb.00265-17] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/29/2017] [Indexed: 12/21/2022] Open
Abstract
Elevated lipid metabolism promotes cancer cell proliferation. Clear cell renal cell carcinoma (ccRCC) is the most common subtype of kidney cancers, characterized by ectopic lipid accumulation. However, the relationship between aberrant lipid metabolism and tumorigenesis in ccRCC is not thoroughly understood. Here, we demonstrate that ring finger protein 20 (RNF20) acts as a tumor suppressor in ccRCC. RNF20 overexpression repressed lipogenesis and cell proliferation by inhibiting sterol regulatory element-binding protein 1c (SREBP1c), and SREBP1 suppression, either by knockdown or by the pharmacological inhibitor betulin, attenuated proliferation and cell cycle progression in ccRCC cells. Notably, SREBP1c regulates cell cycle progression by inducing the expression of pituitary tumor-transforming gene 1 (PTTG1), a novel target gene of SREBP1c. Furthermore, RNF20 overexpression reduced tumor growth and lipid storage in xenografts. In ccRCC patients, RNF20 downregulation and SREBP1 activation are markers of poor prognosis. Therefore, RNF20 suppresses tumorigenesis in ccRCC by inhibiting the SREBP1c-PTTG1 axis.
Collapse
|
46
|
Kim YS, Kim M, Choi MY, Lee DH, Roh GS, Kim HJ, Kang SS, Cho GJ, Park KH, Kim SJ, Yoo JM, Choi WS. Aralia elata (Miq) Seem Extract Decreases O-GlcNAc Transferase Expression and Retinal Cell Death in Diabetic Mice. J Med Food 2017; 20:989-1001. [DOI: 10.1089/jmf.2016.3891] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Yoon Sook Kim
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, South Korea
| | - Minjun Kim
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, South Korea
| | - Mee Young Choi
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, South Korea
| | - Dong Hoon Lee
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, South Korea
| | - Gu Seob Roh
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, South Korea
| | - Hyun Joon Kim
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, South Korea
| | - Sang Soo Kang
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, South Korea
| | - Gyeong Jae Cho
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, South Korea
| | - Ki Hun Park
- Division of Applied Life Science, Institute of Agriculture Life Science, Graduate School of Gyeongsang National University, Jinju, South Korea
| | - Seong-Jae Kim
- Department of Ophthalmology, Gyeongsang National University School of Medicine, Jinju, South Korea
| | - Ji-Myong Yoo
- Department of Ophthalmology, Gyeongsang National University School of Medicine, Jinju, South Korea
| | - Wan Sung Choi
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, South Korea
| |
Collapse
|
47
|
Abstract
Protein modification with the small ubiquitin-related modifier (SUMO) can affect protein function, enzyme activity, protein-protein interactions, protein stability, protein targeting and cellular localization. SUMO influences the function and regulation of metabolic enzymes within pathways, and in some cases targets entire metabolic pathways by affecting the activity of transcription factors or by facilitating the translocation of entire metabolic pathways to subcellular compartments. SUMO modification is also a key component of nutrient- and metabolic-sensing mechanisms that regulate cellular metabolism. In addition to its established roles in maintaining metabolic homeostasis, there is increasing evidence that SUMO is a key factor in facilitating cellular stress responses through the regulation and/or adaptation of the most fundamental metabolic processes, including energy and nucleotide metabolism. This review focuses on the role of SUMO in cellular metabolism and metabolic disease.
Collapse
|
48
|
Lin L, Ding Y, Wang Y, Wang Z, Yin X, Yan G, Zhang L, Yang P, Shen H. Functional lipidomics: Palmitic acid impairs hepatocellular carcinoma development by modulating membrane fluidity and glucose metabolism. Hepatology 2017; 66:432-448. [PMID: 28073184 DOI: 10.1002/hep.29033] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 11/14/2016] [Accepted: 12/26/2016] [Indexed: 01/09/2023]
Abstract
UNLABELLED Lipids are essential cellular components and energy sources of living organisms, and altered lipid composition is increasingly recognized as a signature of cancer. We performed lipidomic analysis in a series of hepatocellular carcinoma (HCC) cells and identified over 1,700 intact lipids originating from three major lipid categories. Comparative lipidomic screening revealed that 93 significantly changed lipids and decreased palmitic acyl (C16:0)-containing glycerophospholipids were positively associated with metastatic abilities of HCC cells. Furthermore, both in vitro and in vivo experiments demonstrated that C16:0 incubation specifically reduced malignant cell proliferation, impaired cell invasiveness, and suppressed tumor growth in mouse xenograft models. Biochemical experiments demonstrated that C16:0 treatment decreased cell membrane fluidity and limited glucose metabolism. A phosphoproteomics approach further revealed such C16:0 incubation attenuated phosphorylation levels of mammalian target of rapamycin (mTOR) and signal transducer and activator of transcription 3 (STAT3) pathway proteins. Multiple reaction monitoring analysis of 443 lipid molecules showed 8 reduced C16:0-containing lipids out of total 10 altered lipids when cancer tissues were compared with adjacent nontumor tissues in a cohort of clinical HCC specimens (P < 0.05). CONCLUSION These data collectively demonstrate the biomedical potential of using altered lipid metabolism as a diagnostic marker for cancerous cells and open an opportunity for treating aggressive HCCs by targeting altered C16:0 metabolism. (Hepatology 2017;66:432-448).
Collapse
Affiliation(s)
- Ling Lin
- Department of Systems Biology for Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Institutes of Biomedical Sciences of Shanghai Medical School, Fudan University, Shanghai, China
| | - Ying Ding
- Department of Systems Biology for Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Shanghai Institute for Food and Drug Control, Shanghai, China
| | - Yi Wang
- Department of Systems Biology for Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Institutes of Biomedical Sciences of Shanghai Medical School, Fudan University, Shanghai, China.,Department of Chemistry, Fudan University, Shanghai, China
| | - Zhenxin Wang
- Department of Systems Biology for Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xuefei Yin
- Department of Systems Biology for Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Department of Chemistry, Fudan University, Shanghai, China
| | - Guoquan Yan
- Department of Chemistry, Fudan University, Shanghai, China
| | - Lei Zhang
- Institutes of Biomedical Sciences of Shanghai Medical School, Fudan University, Shanghai, China
| | - Pengyuan Yang
- Department of Systems Biology for Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Institutes of Biomedical Sciences of Shanghai Medical School, Fudan University, Shanghai, China.,Department of Chemistry, Fudan University, Shanghai, China
| | - Huali Shen
- Department of Systems Biology for Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Institutes of Biomedical Sciences of Shanghai Medical School, Fudan University, Shanghai, China.,Department of Chemistry, Fudan University, Shanghai, China
| |
Collapse
|
49
|
Engelking LJ, Cantoria MJ, Xu Y, Liang G. Developmental and extrahepatic physiological functions of SREBP pathway genes in mice. Semin Cell Dev Biol 2017; 81:98-109. [PMID: 28736205 DOI: 10.1016/j.semcdb.2017.07.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 07/07/2017] [Indexed: 12/17/2022]
Abstract
Sterol regulatory element-binding proteins (SREBPs), master transcriptional regulators of cholesterol and fatty acid synthesis, have been found to contribute to a diverse array of cellular processes. In this review, we focus on genetically engineered mice in which the activities of six components of the SREBP gene pathway, namely SREBP-1, SREBP-2, Scap, Insig-1, Insig-2, or Site-1 protease have been altered through gene knockout or transgenic approaches. In addition to the expected impacts on lipid metabolism, manipulation of these genes in mice is found to affect a wide array of developmental and physiologic processes ranging from interferon signaling in macrophages to synaptic transmission in the brain. The findings reviewed herein provide a blueprint to guide future studies defining the complex interactions between lipid biology and the physiologic processes of many distinct organ systems.
Collapse
Affiliation(s)
- Luke J Engelking
- Departments of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Mary Jo Cantoria
- Departments of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yanchao Xu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Guosheng Liang
- Departments of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
50
|
Rong S, McDonald JG, Engelking LJ. Cholesterol auxotrophy and intolerance to ezetimibe in mice with SREBP-2 deficiency in the intestine. J Lipid Res 2017. [PMID: 28630260 DOI: 10.1194/jlr.m077610] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
SREBP-2 activates transcription of all genes needed for cholesterol biosynthesis. To study SREBP-2 function in the intestine, we generated a mouse model (Vil-BP2-/- ) in which Cre recombinase ablates SREBP-2 in intestinal epithelia. Intestines of Vil-BP2-/- mice had reduced expression of genes required for sterol synthesis, in vivo sterol synthesis rates, and epithelial cholesterol contents. On a cholesterol-free diet, the mice displayed chronic enteropathy with histological abnormalities of both villi and crypts, growth restriction, and reduced survival that was prevented by supplementation of cholesterol in the diet. Likewise, SREBP-2-deficient enteroids required exogenous cholesterol for growth. Blockade of luminal cholesterol uptake into enterocytes with ezetimibe precipitated acutely lethal intestinal damage in Vil-BP2-/- mice, highlighting the critical interplay in the small intestine of sterol absorption via NPC1L1 and sterol synthesis via SREBP-2 in sustaining the intestinal mucosa. These data show that the small intestine requires SREBP-2 to drive cholesterol synthesis that sustains the intestinal epithelia when uptake of cholesterol from the gut lumen is not available, and provide a unique example of cholesterol auxotrophy expressed in an intact, adult mammal.
Collapse
Affiliation(s)
- Shunxing Rong
- Departments of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
| | - Jeffrey G McDonald
- Departments of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046.,Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
| | - Luke J Engelking
- Departments of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046 .,Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046
| |
Collapse
|