1
|
Negi C, Gadara D, Kohoutek J, Bajard L, Spáčil Z, Blaha L. Replacement Flame-Retardant 2-Ethylhexyldiphenyl Phosphate (EHDPP) Disrupts Hepatic Lipidome: Evidence from Human 3D Hepatospheroid Cell Culture. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:2006-2018. [PMID: 36693630 PMCID: PMC9910051 DOI: 10.1021/acs.est.2c03998] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 12/22/2022] [Accepted: 12/22/2022] [Indexed: 05/29/2023]
Abstract
The present study aims to evaluate the effects of repeated exposure to 2-ethylhexyldiphenyl phosphate (EHDPP) on human liver cells. In vitro three-dimensional (3D) hepatospheroid cell culture was utilized to explore the potential mechanisms of EHDPP-mediated metabolic disruption through morphological, transcriptional, and biochemical assays. Lipidomics analysis was performed on the individual hepatospheroids to investigate the effects on intracellular lipid profiles, followed by hepatospheroid morphology, growth, functional parameters, and cytotoxicity evaluation. The possible mechanisms were delineated using the gene-level analysis by assessing the expression of key genes encoding for hepatic lipid metabolism. We revealed that exposure to EHDPP at 1 and 10 μM for 7 days alters the lipid profile of human 3D hepatospheroids. Dysregulation in several lipid classes, including sterol lipids (cholesterol esters), sphingolipids (dihydroceramide, hexosylceramide, ceramide, sphingomyelin), glycerolipids (triglycerides), glycerophospholipids, and fatty acyls, was noted along with alteration in genes including ACAT1, ACAT2, CYP27A1, ABCA1, GPAT2, PNPLA2, PGC1α, and Nrf2. Our study brings a novel insight into the metabolic disrupting effects of EHDPP and demonstrates the utility of hepatospheroids as an in vitro cell culture model complemented with omics technology (e.g., lipidomics) for mechanistic toxicity studies.
Collapse
|
2
|
Kinoo SM, Naidoo P, Singh B, Chuturgoon A, Nagiah S. Human Hepatocyte Nuclear Factors (HNF1 and LXRb) Regulate CYP7A1 in HIV-Infected Black South African Women with Gallstone Disease: A Preliminary Study. Life (Basel) 2023; 13:life13020273. [PMID: 36836631 PMCID: PMC9968087 DOI: 10.3390/life13020273] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/13/2023] [Accepted: 01/14/2023] [Indexed: 01/20/2023] Open
Abstract
Female sex, high estrogen levels, aging, obesity, and dyslipidemia are some of the risk factors associated with gallstone formation. HIV-infected patients on combination antiretroviral therapy (cART) are more prone to hypercholesterolemia. Bile acid synthesis is initiated by cholesterol 7-alpha hydroxylase (CYP7A1) and regulated by hepatocyte nuclear factors (HNF1α, HNF4α, and LXRb). The aim of this study was to evaluate the expression of HNF1α, HNF4α, LXRb, and miRNAs (HNF4α specific: miR-194-5p and miR-122*_1) that regulate CYP7A1 transcription in HIV-infected Black South African women on cART and presenting with gallstones relative to HIV-negative patients with gallstone disease. Females (n = 96) presenting with gallstone disease were stratified based on HIV status. The gene expression of CYP7A1, HNF1α, HNF4α, LXRb, miR-194-5p, and miR-122*_1 was determined using RT-qPCR. Messenger RNA and miRNA levels were reported as fold change expressed as 2-ΔΔCt (RQ min; RQ max). Fold changes >2 and <0.5 were considered significant. HIV-infected females were older in age (p = 0.0267) and displayed higher low-density lipoprotein cholesterol (LDL-c) (p = 0.0419), CYP7A1 [2.078-fold (RQ min: 1.278; RQ max: 3.381)], LXRb [2.595-fold (RQ min: 2.001; RQ max: 3.000)], and HNF1α [3.428 (RQ min: 1.806; RQ max: 6.507] levels. HNF4α [0.642-fold (RQ min: 0.266; RQ max: 1.55)], miR-194-5p [0.527-fold (RQ min: 0.37; RQ max: 0.752)], and miR-122*_1 [0.595-fold (RQ min: 0.332; RQ max: 1.066)] levels were lower in HIV-infected females. In conclusion, HIV-infected women with gallstone disease displayed higher LDL-c levels and increased bile acid synthesis, which was evidenced by the elevated expression of CYP7A1, HNF1α, and LXRb. This could have been further influenced by cART and aging.
Collapse
Affiliation(s)
- Suman Mewa Kinoo
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, College of Health Science, University of KwaZulu Natal, Glenwood, Durban 4041, South Africa
- Discipline of General Surgery, School of Clinical Medicine, College of Health Science, University of KwaZulu Natal, Umbilo, Durban 4001, South Africa
| | - Pragalathan Naidoo
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, College of Health Science, University of KwaZulu Natal, Glenwood, Durban 4041, South Africa
| | - Bhugwan Singh
- Discipline of General Surgery, School of Clinical Medicine, College of Health Science, University of KwaZulu Natal, Umbilo, Durban 4001, South Africa
| | - Anil Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, College of Health Science, University of KwaZulu Natal, Glenwood, Durban 4041, South Africa
- Correspondence: (A.C.); (S.N.)
| | - Savania Nagiah
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, College of Health Science, University of KwaZulu Natal, Glenwood, Durban 4041, South Africa
- Department of Human Biology, Medical School, Faculty of Health Sciences, Nelson Mandela University, Missionvale, Port Elizabeth 6065, South Africa
- Correspondence: (A.C.); (S.N.)
| |
Collapse
|
3
|
Suh JH, Kim KH, Conner ME, Moore DD, Preidis GA. Hepatic PPARα Is Destabilized by SIRT1 Deacetylase in Undernourished Male Mice. Front Nutr 2022; 9:831879. [PMID: 35419389 PMCID: PMC8997242 DOI: 10.3389/fnut.2022.831879] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/10/2022] [Indexed: 11/29/2022] Open
Abstract
The nutrient sensing nuclear receptor peroxisome proliferator-activated receptor-α (PPARα) regulates the host response to short-term fasting by inducing hepatic transcriptional programming of ketogenesis, fatty acid oxidation and transport, and autophagy. This adaptation is ineffective in chronically undernourished individuals, among whom dyslipidemia and hepatic steatosis are common. We recently reported that hepatic PPARα protein is profoundly depleted in male mice undernourished by a low-protein, low-fat diet. Here, we identify PPARα as a deacetylation target of the NAD-dependent deacetylase sirtuin-1 (SIRT1) and link this to the decrease in PPARα protein levels in undernourished liver. Livers from undernourished male mice expressed high levels of SIRT1, with decreased PPARα acetylation and strongly decreased hepatic PPARα protein. In cultured hepatocytes, PPARα protein levels were decreased by transiently transfecting constitutively active SIRT1 or by treating cells with the potent SIRT1 activator resveratrol, while silencing SIRT1 increased PPARα protein levels. SIRT1 expression is correlated with increased PPARα ubiquitination, suggesting that protein loss is due to proteasomal degradation. In accord with these findings, the dramatic loss of hepatic PPARα in undernourished male mice was completely restored by treating mice with the proteasome inhibitor bortezomib. Similarly, treating undernourished mice with the SIRT1 inhibitor selisistat/EX-527 completely restored hepatic PPARα protein. These data suggest that induction of SIRT1 in undernutrition results in hepatic PPARα deacetylation, ubiquitination, and degradation, highlighting a new mechanism that mediates the liver's failed adaptive metabolic responses in chronic undernutrition.
Collapse
Affiliation(s)
- Ji Ho Suh
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States
| | - Kang Ho Kim
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Margaret E Conner
- Department of Molecular Virology and Microbiology, Department of Education, Innovation and Technology, Baylor College of Medicine, Houston, TX, United States
| | - David D Moore
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, United States
| | - Geoffrey A Preidis
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States
| |
Collapse
|
4
|
Role of microRNA-185 in the FoxO1-CYP7A1 mediated regulation of bile acid and cholesterol metabolism: A novel target for drug discovery? Atherosclerosis 2022; 348:53-55. [DOI: 10.1016/j.atherosclerosis.2022.03.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 03/23/2022] [Indexed: 12/31/2022]
|
5
|
MicroRNA-185 modulates CYP7A1 mediated cholesterol-bile acid metabolism through post-transcriptional and post-translational regulation of FoxO1. Atherosclerosis 2022; 348:56-67. [DOI: 10.1016/j.atherosclerosis.2022.03.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/22/2022] [Accepted: 03/03/2022] [Indexed: 12/22/2022]
|
6
|
Lu D, Liu Y, Luo Y, Zhao J, Feng C, Xue L, Xu J, Wang Q, Yan T, Xiao P, Krausz KW, Gonzalez FJ, Xie C. Intestinal farnesoid X receptor signaling controls hepatic fatty acid oxidation. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159089. [PMID: 34856412 PMCID: PMC8864892 DOI: 10.1016/j.bbalip.2021.159089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 02/03/2023]
Abstract
In addition to maintaining bile acid, cholesterol and glucose homeostasis, farnesoid X receptor (FXR) also regulates fatty acid β-oxidation (FAO). To explore the different roles of hepatic and intestinal FXR in liver FAO, FAO-associated metabolites, including acylcarnitines and fatty acids, and FXR target gene mRNAs were profiled using an integrated metabolomic and transcriptomic analysis in control (Fxrfl/fl), liver-specific Fxr-null (FxrΔHep) and intestine-specific Fxr-null (FxrΔIE) mice, treated either with the FXR agonist obeticholic acid (OCA) or vehicle (VEH). Activation of FXR by OCA treatment significantly increased fatty acyl-CoA hydrolysis (Acot1) and decreased FAO-associated mRNAs in Fxrfl/fl mice, resulting in reduced levels of total acylcarnitines and relative accumulation of long/medium chain acylcarnitines and fatty acids in liver. FxrΔHep mice responded to OCA treatment in a manner similar to Fxrfl/fl mice while FxrΔIE mice responded differently, thus illustrating that intestinal FXR plays a critical role in the regulation of hepatic FAO. A significant negative-correlation between intestinal FXR-FGF15 and hepatic CREB-PGC1A pathways was observed after both VEH and OCA treatment, suggesting that OCA-induced activation of the intestinal FXR-FGF15 axis downregulates hepatic PGC1α signaling via inactivation of hepatic CREB, thus repressing FAO. This mechanism was confirmed in experiments based on human recombinant FGF19 treatment and intestinal Fgf15-null mice. This study revealed an important role for the intestinal FXR-FGF15 pathway in hepatic FAO repression.
Collapse
Affiliation(s)
- Dasheng Lu
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America; Shanghai Municipal Center for Disease Control and Prevention, State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, Shanghai, PR China
| | - Yameng Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China
| | - Yuhong Luo
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Jie Zhao
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Chao Feng
- Shanghai Municipal Center for Disease Control and Prevention, State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, Shanghai, PR China
| | - Liming Xue
- Shanghai Municipal Center for Disease Control and Prevention, State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, Shanghai, PR China
| | - Jiale Xu
- Shanghai Municipal Center for Disease Control and Prevention, State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, Shanghai, PR China
| | - Qiong Wang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Tingting Yan
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Ping Xiao
- Shanghai Municipal Center for Disease Control and Prevention, State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, Shanghai, PR China
| | - Kristopher W Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America.
| | - Cen Xie
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China.
| |
Collapse
|
7
|
Shen Z, Zhu W, Du L. Analysis of Gene Expression Profiles in the Liver of Rats With Intrauterine Growth Retardation. Front Pediatr 2022; 10:801544. [PMID: 35321016 PMCID: PMC8934861 DOI: 10.3389/fped.2022.801544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Intrauterine growth restriction (IUGR) is highly associated with fetal as well as neonatal morbidity, mortality, and an increased risk metabolic disease development later in life. The mechanism involved in the increased risk has not been established. We compared differentially expressed genes between the liver of appropriate for gestational age (AGA) and IUGR rat models and identified their effects on molecular pathways involved in the metabolic syndrome. METHODS We extracted RNA from the liver of IUGR and AGA rats and profiled gene expression by microarray analysis. GO function and KEGG pathway enrichment analyses were conducted using the Search Tool for the Retrieval of Interacting Genes database. Then, the Cytoscape software was used to visualize regulatory interaction networks of IUGR-related genes. The results were further verified via quantitative reverse transcriptase PCR analysis. RESULTS In this study, 815 genes were found to be markedly differentially expressed (fold-change >1.5, p < 0.05) between IUGR and AGA, with 347 genes elevated and 468 suppressed in IUGR, relative to AGA. Enrichment and protein-protein interaction network analyses of target genes revealed that core genes including Ppargc1a, Prkaa2, Slc2a1, Rxrg, and Gcgr, and pathways, including the PPAR signaling pathway and FoxO signaling pathway, had a potential association with metabolic syndrome development in IUGR. We also confirmed that at the mRNA level, five genes involved in glycometabolism were differentially expressed between IUGR and AGA. CONCLUSION Our findings elucidate on differential gene expression profiles in IUGR and AGA. Moreover, they elucidate on the pathogenesis of IUGR-associated metabolic syndromes. The suggested candidates are potential biomarkers and eventually intended to treat them appropriately.
Collapse
Affiliation(s)
- Zheng Shen
- Department of Clinical Laboratory, Zhejiang University School of Medicine Children's Hospital, Hangzhou, China.,National Clinical Research Center for Child Health, Hangzhou, China
| | - Weifen Zhu
- Department of Endocrinology, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Hangzhou, China
| | - Lizhong Du
- National Clinical Research Center for Child Health, Hangzhou, China.,Department of Neonatology, Zhejiang University School of Medicine Children's Hospital, Hangzhou, China
| |
Collapse
|
8
|
Mulberry Leaf and Radix Astragali Regulates Differentially Expressed Genes and Proteins in the Streptozotocin-Induced Diabetic Mice Liver. Processes (Basel) 2021. [DOI: 10.3390/pr9111898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
As a chronic non-infectious disease, severely affecting human quality and health of life, diabetes mellitus (DM) and its complications have gradually developed into a major global public health problem. Mulberry Leaf and Radix Astragali have been used as a traditional medicinal formulation in diabetic patients for a long time, whose combination is usually found in traditional Chinese medicine prescriptions. However, due to the unclear synergistic mechanism of them for DM, the changes of differential genes and proteins in the liver tissue of streptozotocin-induced diabetic mice were analyzed, and then the potential synergistic mechanism of them in anti-diabetes was investigated in our research. Compared with the diabetic model group, there were 699 differentially expressed genes and 169 differentially expressed proteins in the Mulberry Leaf and Radix Astragali treated group, and there were 35 common specific genes both in the transcriptome and the proteome. These common genes participated mainly in the pathways, such as retinol metabolism, steroid hormone biosynthesis, and arachidonic acid metabolism. Quantitative real-time PCR() and Western blot results speculated that the synergistic effect on anti-diabetes was mainly through regulating the expression of Tap1, Ncoa4, and Alas2, by down-regulating Fabp2 and Hmox1 and up-regulating Hmgcr, Cyp7a1. All these genes would affect bile acid secretion, alleviate the occurrence of iron death, promote the metabolism and synthesis of glycolipid substances, and ultimately maintain the body’s glucose homeostasis.
Collapse
|
9
|
Nguyen JT, Riessen R, Zhang T, Kieffer C, Anakk S. Deletion of Intestinal SHP Impairs Short-term Response to Cholic Acid Challenge in Male Mice. Endocrinology 2021; 162:6189092. [PMID: 33769482 PMCID: PMC8256632 DOI: 10.1210/endocr/bqab063] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Indexed: 02/07/2023]
Abstract
Small heterodimer partner (SHP) is a crucial regulator of bile acid (BA) transport and synthesis; however, its intestine-specific role is not fully understood. Here, we report that male intestine-specific Shp knockout (IShpKO) mice exhibit higher intestinal BA but not hepatic or serum BA levels compared with the f/f Shp animals when challenged with an acute (5-day) 1% cholic acid (CA) diet. We also found that BA synthetic genes Cyp7a1 and Cyp8b1 are not repressed to the same extent in IShpKO compared with control mice post-CA challenge. Loss of intestinal SHP did not alter Fxrα messenger RNA (mRNA) but increased Asbt (BA ileal uptake transporter) and Ostα (BA ileal efflux transporter) expression even under chow-fed conditions. Surprisingly, the acute CA diet in IShpKO did not elicit the expected induction of Fgf15 but was able to maintain the suppression of Asbt, and Ostα/β mRNA levels. At the protein level, apical sodium-dependent bile acid transporter (ASBT) was downregulated, while organic solute transporter-α/β (OSTα/β) expression was induced and maintained regardless of diet. Examination of ileal histology in IShpKO mice challenged with acute CA diet revealed reduced villi length and goblet cell numbers. However, no difference in villi length, and the expression of BA regulator and transporter genes, was seen between f/f Shp and IShpKO animals after a chronic (14-day) CA diet, suggesting a potential adaptive response. We found the upregulation of the Pparα-Ugt axis after 14 days of CA diet may reduce the BA burden and compensate for the ileal SHP function. Thus, our study reveals that ileal SHP expression contributes to both overall intestinal structure and BA homeostasis.
Collapse
Affiliation(s)
- James T Nguyen
- Department of Molecular & Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Ryan Riessen
- Department of Molecular & Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Tongyu Zhang
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Collin Kieffer
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Sayeepriyadarshini Anakk
- Department of Molecular & Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Correspondence:Sayeepriyadarshini Anakk, Department of Molecular & Integrative Physiology and Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 450 Medical Science Building, 506 South Matthews Avenue, Urbana, IL 61801, USA. E-mail:
| |
Collapse
|
10
|
PGC1s and Beyond: Disentangling the Complex Regulation of Mitochondrial and Cellular Metabolism. Int J Mol Sci 2021; 22:ijms22136913. [PMID: 34199142 PMCID: PMC8268830 DOI: 10.3390/ijms22136913] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 02/07/2023] Open
Abstract
Metabolism is the central engine of living organisms as it provides energy and building blocks for many essential components of each cell, which are required for specific functions in different tissues. Mitochondria are the main site for energy production in living organisms and they also provide intermediate metabolites required for the synthesis of other biologically relevant molecules. Such cellular processes are finely tuned at different levels, including allosteric regulation, posttranslational modifications, and transcription of genes encoding key proteins in metabolic pathways. Peroxisome proliferator activated receptor γ coactivator 1 (PGC1) proteins are transcriptional coactivators involved in the regulation of many cellular processes, mostly ascribable to metabolic pathways. Here, we will discuss some aspects of the cellular processes regulated by PGC1s, bringing up some examples of their role in mitochondrial and cellular metabolism, and how metabolic regulation in mitochondria by members of the PGC1 family affects the immune system. We will analyze how PGC1 proteins are regulated at the transcriptional and posttranslational level and will also examine other regulators of mitochondrial metabolism and the related cellular functions, considering approaches to identify novel mitochondrial regulators and their role in physiology and disease. Finally, we will analyze possible therapeutical perspectives currently under assessment that are applicable to different disease states.
Collapse
|
11
|
Li H, Yu XH, Ou X, Ouyang XP, Tang CK. Hepatic cholesterol transport and its role in non-alcoholic fatty liver disease and atherosclerosis. Prog Lipid Res 2021; 83:101109. [PMID: 34097928 DOI: 10.1016/j.plipres.2021.101109] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a quickly emerging global health problem representing the most common chronic liver disease in the world. Atherosclerotic cardiovascular disease represents the leading cause of mortality in NAFLD patients. Cholesterol metabolism has a crucial role in the pathogenesis of both NAFLD and atherosclerosis. The liver is the major organ for cholesterol metabolism. Abnormal hepatic cholesterol metabolism not only leads to NAFLD but also drives the development of atherosclerotic dyslipidemia. The cholesterol level in hepatocytes reflects the dynamic balance between endogenous synthesis, uptake, esterification, and export, a process in which cholesterol is converted to neutral cholesteryl esters either for storage in cytosolic lipid droplets or for secretion as a major constituent of plasma lipoproteins, including very-low-density lipoproteins, chylomicrons, high-density lipoproteins, and low-density lipoproteins. In this review, we describe decades of research aimed at identifying key molecules and cellular players involved in each main aspect of hepatic cholesterol metabolism. Furthermore, we summarize the recent advances regarding the biological processes of hepatic cholesterol transport and its role in NAFLD and atherosclerosis.
Collapse
Affiliation(s)
- Heng Li
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Xiao-Hua Yu
- Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 460106, China
| | - Xiang Ou
- Department of Endocrinology, the First Hospital of Changsha, Changsha, Hunan 410005, China
| | - Xin-Ping Ouyang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China.
| | - Chao-Ke Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
12
|
Role of osteopontin in diet-induced brown gallstone formation in rats. Chin Med J (Engl) 2021; 134:1093-1100. [PMID: 33883409 PMCID: PMC8116003 DOI: 10.1097/cm9.0000000000001519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background: Although osteopontin (OPN) is expressed in the liver and pigment gallstones of patients with hepatolithiasis, its role in pigment gallstone formation remains unclear. This study aimed to explore the function of OPN in pigment gallstone formation. Methods: Rats were fed a chow diet (CD) or lithogenic diet (LD) for 10 consecutive weeks; blocking tests were then performed using an OPN antibody (OPN-Ab). Incidence of gallstones and levels of several bile components, OPN, tumor necrosis factor alpha (TNF-α), and cholesterol 7 alpha-hydroxylase (CYP7A1) were analyzed. To determine TNF-α expression in hepatic macrophages and both CYP7A1 and bile acid (BA) expression in liver cells, recombinant rat OPN and recombinant rat TNF-α were used to treat rat hepatic macrophages and rat liver cells, respectively. Chi-square or Fisher exact tests were used to analyze qualitative data, Student t-test or one-way analysis of variance were used to analyze qualitative data. Results: Incidence of gallstones was higher in LD-fed rats than in CD-fed rats (80% vs. 10%, P < 0.05). BA content significantly decreased in bile (t = −36.08, P < 0.01) and liver tissue (t = −16.16, P < 0.01) of LD-fed rats. Both hepatic OPN protein expression (t = 9.78, P < 0.01) and TNF-α level (t = 8.83, P < 0.01) distinctly increased in the LD group; what's more, CYP7A1 mRNA and protein levels (t = −12.35, P < 0.01) were markedly down-regulated in the LD group. Following OPN-Ab pretreatment, gallstone formation decreased (85% vs. 25%, χ2 = 14.55, P < 0.01), liver TNF-α expression (F = 20.36, P < 0.01) was down-regulated in the LD group, and CYP7A1 expression (F = 17.51, P < 0.01) was up-regulated. Through CD44 and integrin receptors, OPN promoted TNF-α production in macrophage (F = 1041, P < 0.01), which suppressed CYP7A1 expression (F = 48.08, P < 0.01) and reduced liver BA synthesis (F = 119.4, P < 0.01). Conclusions: We provide novel evidence of OPN involvement in pigmented gallstone pathogenesis in rats.
Collapse
|
13
|
Solares I, Izquierdo-Sánchez L, Morales-Conejo M, Jericó D, Castelbón FJ, Córdoba KM, Sampedro A, Lumbreras C, Moreno-Aliaga MJ, Enríquez de Salamanca R, Berraondo P, Fontanellas A. High Prevalence of Insulin Resistance in Asymptomatic Patients with Acute Intermittent Porphyria and Liver-Targeted Insulin as a Novel Therapeutic Approach. Biomedicines 2021; 9:255. [PMID: 33807619 PMCID: PMC8002016 DOI: 10.3390/biomedicines9030255] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/22/2021] [Accepted: 02/27/2021] [Indexed: 02/08/2023] Open
Abstract
Acute porphyria attacks are associated with the strong up-regulation of hepatic heme synthesis and over-production of neurotoxic heme precursors. First-line therapy is based on carbohydrate loading. However, altered glucose homeostasis could affect its efficacy. Our first aim was to investigate the prevalence of insulin resistance (IR) in an observational case-control study including 44 Spanish patients with acute intermittent porphyria (AIP) and 55 age-, gender- and BMI-matched control volunteers. Eight patients (18.2%) and one control (2.3%, p = 0.01) showed a high HOMA-IR index (cut-off ≥ 3.4). Patients with IR and hyperinsulinemia showed clinically stable disease. Thus, the second aim was to evaluate the effect of the co-administration of glucose and a fast-acting or new liver-targeted insulin (the fusion protein of insulin and apolipoprotein A-I, Ins-ApoAI) in AIP mice. The combination of glucose and the Ins-ApoAI promoted partial but sustained protection against hepatic heme synthesis up-regulation compared with glucose alone or co-injected with fast-acting insulin. In a prevention study, Ins-ApoAI improved symptoms associated with a phenobarbital-induced attack but maintained high porphyrin precursor excretion, probably due to the induction of hepatic mitochondrial biogenesis mediated by apolipoprotein A-I. In conclusion, a high prevalence of IR and hyperinsulinemia was observed in patients with AIP. The experimental data provide proof-of-concept for liver-targeted insulin as a way of enhancing glucose therapy for AIP.
Collapse
Affiliation(s)
- Isabel Solares
- Reference Center for Inherited Metabolic Disease-MetabERN, Department of Internal Medicine, University Hospital 12 de Octubre, UCM, 28041 Madrid, Spain; (I.S.); (M.M.-C.); (F.J.C.); (C.L.); (R.E.d.S.)
| | - Laura Izquierdo-Sánchez
- Hepatology Program, Cima Universidad de Navarra, 31008 Pamplona, Spain; (L.I.-S.); (D.J.); (K.M.C.); (A.S.)
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), 20014 San Sebastian, Spain
| | - Montserrat Morales-Conejo
- Reference Center for Inherited Metabolic Disease-MetabERN, Department of Internal Medicine, University Hospital 12 de Octubre, UCM, 28041 Madrid, Spain; (I.S.); (M.M.-C.); (F.J.C.); (C.L.); (R.E.d.S.)
- Grupo de Enfermedades Mitocondriales y Neuromusculares, Instituto de Investigación Hospital 12 de Octubre (i+12), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Daniel Jericó
- Hepatology Program, Cima Universidad de Navarra, 31008 Pamplona, Spain; (L.I.-S.); (D.J.); (K.M.C.); (A.S.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (M.J.M.-A.); (P.B.)
| | - Francisco Javier Castelbón
- Reference Center for Inherited Metabolic Disease-MetabERN, Department of Internal Medicine, University Hospital 12 de Octubre, UCM, 28041 Madrid, Spain; (I.S.); (M.M.-C.); (F.J.C.); (C.L.); (R.E.d.S.)
| | - Karol Marcela Córdoba
- Hepatology Program, Cima Universidad de Navarra, 31008 Pamplona, Spain; (L.I.-S.); (D.J.); (K.M.C.); (A.S.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (M.J.M.-A.); (P.B.)
| | - Ana Sampedro
- Hepatology Program, Cima Universidad de Navarra, 31008 Pamplona, Spain; (L.I.-S.); (D.J.); (K.M.C.); (A.S.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (M.J.M.-A.); (P.B.)
| | - Carlos Lumbreras
- Reference Center for Inherited Metabolic Disease-MetabERN, Department of Internal Medicine, University Hospital 12 de Octubre, UCM, 28041 Madrid, Spain; (I.S.); (M.M.-C.); (F.J.C.); (C.L.); (R.E.d.S.)
| | - María Jesús Moreno-Aliaga
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (M.J.M.-A.); (P.B.)
- Center for Nutrition Research and Department of Nutrition, Food Science and Physiology, University of Navarra, 31008 Pamplona, Spain
- CIBERobn Physiopathology of Obesity and Nutrition, Carlos III Health Institute, 28029 Madrid, Spain
| | - Rafael Enríquez de Salamanca
- Reference Center for Inherited Metabolic Disease-MetabERN, Department of Internal Medicine, University Hospital 12 de Octubre, UCM, 28041 Madrid, Spain; (I.S.); (M.M.-C.); (F.J.C.); (C.L.); (R.E.d.S.)
| | - Pedro Berraondo
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (M.J.M.-A.); (P.B.)
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERonc, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Antonio Fontanellas
- Hepatology Program, Cima Universidad de Navarra, 31008 Pamplona, Spain; (L.I.-S.); (D.J.); (K.M.C.); (A.S.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (M.J.M.-A.); (P.B.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
14
|
López-Soldado I, Guinovart JJ, Duran J. Increasing hepatic glycogen moderates the diabetic phenotype in insulin-deficient Akita mice. J Biol Chem 2021; 296:100498. [PMID: 33667544 PMCID: PMC8027280 DOI: 10.1016/j.jbc.2021.100498] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/23/2021] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
Hepatic glycogen metabolism is impaired in diabetes. We previously demonstrated that strategies to increase liver glycogen content in a high-fat-diet mouse model of obesity and insulin resistance led to a reduction in food intake and ameliorated obesity and glucose tolerance. These effects were accompanied by a decrease in insulin levels, but whether this decrease contributed to the phenotype observed in this animal was unclear. Here we sought to evaluate this aspect directly, by examining the long-term effects of increasing liver glycogen in an animal model of insulin-deficient and monogenic diabetes, namely the Akita mouse, which is characterized by reduced insulin production. We crossed Akita mice with animals overexpressing protein targeting to glycogen (PTG) in the liver to generate Akita mice with increased liver glycogen content (Akita-PTGOE). Akita-PTGOE animals showed lower glycemia, lower food intake, and decreased water consumption and urine output compared with Akita mice. Furthermore, Akita-PTGOE mice showed a restoration of the hepatic energy state and a normalization of gluconeogenesis and glycolysis back to nondiabetic levels. Moreover, hepatic lipogenesis, which is reduced in Akita mice, was reverted in Akita-PTGOE animals. These results demonstrate that strategies to increase liver glycogen content lead to the long-term reduction of the diabetic phenotype, independently of circulating insulin.
Collapse
Affiliation(s)
- Iliana López-Soldado
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain.
| | - Joan J Guinovart
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain; Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain
| | - Jordi Duran
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| |
Collapse
|
15
|
Pedersen AF, Meyer DN, Petriv AMV, Soto AL, Shields JN, Akemann C, Baker BB, Tsou WL, Zhang Y, Baker TR. Nanoplastics impact the zebrafish (Danio rerio) transcriptome: Associated developmental and neurobehavioral consequences. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 266:115090. [PMID: 32693326 PMCID: PMC7492438 DOI: 10.1016/j.envpol.2020.115090] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/05/2020] [Accepted: 06/22/2020] [Indexed: 05/20/2023]
Abstract
Microplastics (MPs) are a ubiquitous pollutant detected not only in marine and freshwater bodies, but also in tap and bottled water worldwide. While MPs have been extensively studied, the toxicity of their smaller counterpart, nanoplastics (NPs), is not well documented. Despite likely large-scale human and animal exposure to NPs, the associated health risks remain unclear, especially during early developmental stages. To address this, we investigated the health impacts of exposures to both 50 and 200 nm polystyrene NPs in larval zebrafish. From 6 to 120 h post-fertilization (hpf), developing zebrafish were exposed to a range of fluorescent NPs (10-10,000 parts per billion). Dose-dependent increases in accumulation were identified in exposed larval fish, potentially coinciding with an altered behavioral response as evidenced through swimming hyperactivity. Notably, exposures did not impact mortality, hatching rate, or deformities; however, transcriptomic analysis suggests neurodegeneration and motor dysfunction at both high and low concentrations. Furthermore, results of this study suggest that NPs can accumulate in the tissues of larval zebrafish, alter their transcriptome, and affect behavior and physiology, potentially decreasing organismal fitness in contaminated ecosystems. The uniquely broad scale of this study during a critical window of development provides crucial multidimensional characterization of NP impacts on human and animal health.
Collapse
Affiliation(s)
- Adam F Pedersen
- Institute of Environmental Health Sciences, Wayne State University, 6135 Woodward Ave, Detroit, MI, 48202, USA
| | - Danielle N Meyer
- Institute of Environmental Health Sciences, Wayne State University, 6135 Woodward Ave, Detroit, MI, 48202, USA; Department of Pharmacology - School of Medicine, Wayne State University, 540 E Canfield, Detroit, MI, 28201, USA
| | - Anna-Maria V Petriv
- Institute of Environmental Health Sciences, Wayne State University, 6135 Woodward Ave, Detroit, MI, 48202, USA
| | - Abraham L Soto
- Institute of Environmental Health Sciences, Wayne State University, 6135 Woodward Ave, Detroit, MI, 48202, USA
| | - Jeremiah N Shields
- Institute of Environmental Health Sciences, Wayne State University, 6135 Woodward Ave, Detroit, MI, 48202, USA
| | - Camille Akemann
- Institute of Environmental Health Sciences, Wayne State University, 6135 Woodward Ave, Detroit, MI, 48202, USA; Department of Pharmacology - School of Medicine, Wayne State University, 540 E Canfield, Detroit, MI, 28201, USA
| | - Bridget B Baker
- Institute of Environmental Health Sciences, Wayne State University, 6135 Woodward Ave, Detroit, MI, 48202, USA
| | - Wei-Ling Tsou
- Department of Pharmacology - School of Medicine, Wayne State University, 540 E Canfield, Detroit, MI, 28201, USA
| | - Yongli Zhang
- College of Engineering, Wayne State University, 5050 Anthony Wayne Dr, Detroit, MI, 28201, USA
| | - Tracie R Baker
- Institute of Environmental Health Sciences, Wayne State University, 6135 Woodward Ave, Detroit, MI, 48202, USA; Department of Pharmacology - School of Medicine, Wayne State University, 540 E Canfield, Detroit, MI, 28201, USA.
| |
Collapse
|
16
|
Bae-Gartz I, Kasper P, Großmann N, Breuer S, Janoschek R, Kretschmer T, Appel S, Schmitz L, Vohlen C, Quaas A, Schweiger MR, Grimm C, Fischer A, Ferrari N, Graf C, Frese CK, Lang S, Demir M, Schramm C, Fink G, Goeser T, Dötsch J, Hucklenbruch-Rother E. Maternal exercise conveys protection against NAFLD in the offspring via hepatic metabolic programming. Sci Rep 2020; 10:15424. [PMID: 32963289 PMCID: PMC7508970 DOI: 10.1038/s41598-020-72022-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 05/11/2020] [Indexed: 12/13/2022] Open
Abstract
Maternal exercise (ME) during pregnancy has been shown to improve metabolic health in offspring and confers protection against the development of non-alcoholic fatty liver disease (NAFLD). However, its underlying mechanism are still poorly understood, and it remains unclear whether protective effects on hepatic metabolism are already seen in the offspring early life. This study aimed at determining the effects of ME during pregnancy on offspring body composition and development of NAFLD while focusing on proteomic-based analysis of the hepatic energy metabolism during developmental organ programming in early life. Under an obesogenic high-fat diet (HFD), male offspring of exercised C57BL/6J-mouse dams were protected from body weight gain and NAFLD in adulthood (postnatal day (P) 112). This was associated with a significant activation of hepatic AMP-activated protein kinase (AMPK), peroxisome proliferator-activated receptor alpha (PPARα) and PPAR coactivator-1 alpha (PGC1α) signaling with reduced hepatic lipogenesis and increased hepatic β-oxidation at organ programming peak in early life (P21). Concomitant proteomic analysis revealed a characteristic hepatic expression pattern in offspring as a result of ME with the most prominent impact on Cholesterol 7 alpha-hydroxylase (CYP7A1). Thus, ME may offer protection against offspring HFD-induced NAFLD by shaping hepatic proteomics signature and metabolism in early life. The results highlight the potential of exercise during pregnancy for preventing the early origins of NAFLD.
Collapse
Affiliation(s)
- Inga Bae-Gartz
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Robert-Koch Str. 16, Building 44a, 50931, Cologne, Germany.
| | - Philipp Kasper
- Department of Gastroenterology and Hepatology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Nora Großmann
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Robert-Koch Str. 16, Building 44a, 50931, Cologne, Germany
| | - Saida Breuer
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Robert-Koch Str. 16, Building 44a, 50931, Cologne, Germany
| | - Ruth Janoschek
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Robert-Koch Str. 16, Building 44a, 50931, Cologne, Germany
| | - Tobias Kretschmer
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Robert-Koch Str. 16, Building 44a, 50931, Cologne, Germany
| | - Sarah Appel
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Robert-Koch Str. 16, Building 44a, 50931, Cologne, Germany
| | - Lisa Schmitz
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Robert-Koch Str. 16, Building 44a, 50931, Cologne, Germany
| | - Christina Vohlen
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Robert-Koch Str. 16, Building 44a, 50931, Cologne, Germany
| | - Alexander Quaas
- Department of Pathology, University Hospital of Cologne, Cologne, Germany
| | - Michal R Schweiger
- Translational Epigenetics and Tumor Genetic, University Hospital of Cologne, Cologne, Germany
| | - Christina Grimm
- Translational Epigenetics and Tumor Genetic, University Hospital of Cologne, Cologne, Germany
| | | | - Nina Ferrari
- Cologne Center for Prevention in Childhood and Youth / Heart Center Cologne, University Hospital of Cologne, Cologne, Germany.,Institute of Movement and Neuroscience, Department of Movement and Health Promotion, German Sport University, Cologne, Germany
| | - Christine Graf
- Institute of Movement and Neuroscience, Department of Movement and Health Promotion, German Sport University, Cologne, Germany
| | - Christian K Frese
- Proteomics Core Facility, CECAD Research Center, University Hospital of Cologne, Cologne, Germany.,Max-Planck-Unit for the Science of Pathogens, Charité University Medicine Berlin, Berlin, Germany
| | - Sonja Lang
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Münevver Demir
- Department of Hepatology and Gastroenterology, Charité Campus Mitte and Campus Virchow Clinic, Charité University Medicine Berlin, Berlin, Germany
| | - Christoph Schramm
- Department of Gastroenterology and Hepatology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Gregor Fink
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Robert-Koch Str. 16, Building 44a, 50931, Cologne, Germany
| | - Tobias Goeser
- Department of Gastroenterology and Hepatology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Jörg Dötsch
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Robert-Koch Str. 16, Building 44a, 50931, Cologne, Germany
| | - Eva Hucklenbruch-Rother
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Robert-Koch Str. 16, Building 44a, 50931, Cologne, Germany
| |
Collapse
|
17
|
Wang Y, Gunewardena S, Li F, Matye DJ, Chen C, Chao X, Jung T, Zhang Y, Czerwiński M, Ni HM, Ding WX, Li T. An FGF15/19-TFEB regulatory loop controls hepatic cholesterol and bile acid homeostasis. Nat Commun 2020; 11:3612. [PMID: 32681035 PMCID: PMC7368063 DOI: 10.1038/s41467-020-17363-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 06/26/2020] [Indexed: 12/15/2022] Open
Abstract
Bile acid synthesis plays a key role in regulating whole body cholesterol homeostasis. Transcriptional factor EB (TFEB) is a nutrient and stress-sensing transcriptional factor that promotes lysosomal biogenesis. Here we report a role of TFEB in regulating hepatic bile acid synthesis. We show that TFEB induces cholesterol 7α-hydroxylase (CYP7A1) in human hepatocytes and mouse livers and prevents hepatic cholesterol accumulation and hypercholesterolemia in Western diet-fed mice. Furthermore, we find that cholesterol-induced lysosomal stress feed-forward activates TFEB via promoting TFEB nuclear translocation, while bile acid-induced fibroblast growth factor 19 (FGF19), acting via mTOR/ERK signaling and TFEB phosphorylation, feedback inhibits TFEB nuclear translocation in hepatocytes. Consistently, blocking intestinal bile acid uptake by an apical sodium-bile acid transporter (ASBT) inhibitor decreases ileal FGF15, enhances hepatic TFEB nuclear localization and improves cholesterol homeostasis in Western diet-fed mice. This study has identified a TFEB-mediated gut-liver signaling axis that regulates hepatic cholesterol and bile acid homeostasis.
Collapse
Affiliation(s)
- Yifeng Wang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Sumedha Gunewardena
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Feng Li
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - David J Matye
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Cheng Chen
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Xiaojuan Chao
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Taeyoon Jung
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Yuxia Zhang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | | | - Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Tiangang Li
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
18
|
Mehmel M, Jovanović N, Spitz U. Nicotinamide Riboside-The Current State of Research and Therapeutic Uses. Nutrients 2020; 12:E1616. [PMID: 32486488 PMCID: PMC7352172 DOI: 10.3390/nu12061616] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 12/19/2022] Open
Abstract
Nicotinamide riboside (NR) has recently become one of the most studied nicotinamide adenine dinucleotide (NAD+) precursors, due to its numerous potential health benefits mediated via elevated NAD+ content in the body. NAD+ is an essential coenzyme that plays important roles in various metabolic pathways and increasing its overall content has been confirmed as a valuable strategy for treating a wide variety of pathophysiological conditions. Accumulating evidence on NRs' health benefits has validated its efficiency across numerous animal and human studies for the treatment of a number of cardiovascular, neurodegenerative, and metabolic disorders. As the prevalence and morbidity of these conditions increases in modern society, the great necessity has arisen for a rapid translation of NR to therapeutic use and further establishment of its availability as a nutritional supplement. Here, we summarize currently available data on NR effects on metabolism, and several neurodegenerative and cardiovascular disorders, through to its application as a treatment for specific pathophysiological conditions. In addition, we have reviewed newly published research on the application of NR as a potential therapy against infections with several pathogens, including SARS-CoV-2. Additionally, to support rapid NR translation to therapeutics, the challenges related to its bioavailability and safety are addressed, together with the advantages of NR to other NAD+ precursors.
Collapse
Affiliation(s)
- Mario Mehmel
- Biosynth Carbosynth, Rietlistrasse 4, 9422 Staad, Switzerland;
| | - Nina Jovanović
- Faculty of Biology, Department of Biochemistry and Molecular Biology, Institute of Physiology and Biochemistry, University of Belgrade, Studentski Trg 1, 11000 Belgrade, Serbia;
| | - Urs Spitz
- Biosynth Carbosynth, Axis House, High Street, Compton, Berkshire RG20 6NL, UK
| |
Collapse
|
19
|
Piccinin E, Arconzo M, Graziano G, Vacca M, Peres C, Bellafante E, Villani G, Moschetta A. Hepatic microRNA Expression by PGC-1α and PGC-1β in the Mouse. Int J Mol Sci 2019; 20:ijms20225735. [PMID: 31731670 PMCID: PMC6888418 DOI: 10.3390/ijms20225735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023] Open
Abstract
The fine-tuning of liver metabolism is essential to maintain the whole-body homeostasis and to prevent the onset of diseases. The peroxisome proliferator-activated receptor-γ coactivators (PGC-1s) are transcriptional key players of liver metabolism, able to regulate mitochondrial function, gluconeogenesis and lipid metabolism. Their activity is accurately modulated by post-translational modifications. Here, we showed that specific PGC-1s expression can lead to the upregulation of different microRNAs widely implicated in liver physiology and diseases development and progression, thus offering a new layer of complexity in the control of hepatic metabolism.
Collapse
Affiliation(s)
- Elena Piccinin
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy or
| | - Maria Arconzo
- INBB, National Institute for Biostuctures and Biosystems, 00136 Rome, Italy; (M.A.); (G.G.); (C.P.)
| | - Giusi Graziano
- INBB, National Institute for Biostuctures and Biosystems, 00136 Rome, Italy; (M.A.); (G.G.); (C.P.)
| | - Michele Vacca
- Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK;
| | - Claudia Peres
- INBB, National Institute for Biostuctures and Biosystems, 00136 Rome, Italy; (M.A.); (G.G.); (C.P.)
| | - Elena Bellafante
- Fondazione Mario Negri Sud, Santa Maria Imbaro, 66030 Chieti, Italy;
| | - Gaetano Villani
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, “Aldo Moro” University of Bari, 70124 Bari, Italy;
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy or
- INBB, National Institute for Biostuctures and Biosystems, 00136 Rome, Italy; (M.A.); (G.G.); (C.P.)
- Correspondence: or ; Tel.: +39-080-559-3262
| |
Collapse
|
20
|
Bhatta P, Bermano G, Williams HC, Knott RM. Meta-analysis demonstrates Gly482Ser variant of PPARGC1A is associated with components of metabolic syndrome within Asian populations. Genomics 2019; 112:1795-1803. [PMID: 31678594 DOI: 10.1016/j.ygeno.2019.10.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/11/2019] [Accepted: 10/17/2019] [Indexed: 11/18/2022]
Abstract
AIM To determine the association of peroxisome proliferator activated receptor gamma coactivator 1 Gly482Ser variant with components of metabolic syndrome. MATERIALS AND METHODS A systematic search was carried out using Web of Science, PubMed, EMBASE and the Cochrane library using the key words: Peroxisome proliferator activator receptor gamma coactivator 1, PPARGC1A, PGC-1, PGC-1alpha, and PGC1alpha alone or with polymorphism, Gly482Ser and rs8192678. RESULTS Data from 19 articles generated 28 separate data sets. Under the recessive model fasting plasma glucose was significantly lower in AA genotypes when compared to GG + GA in the total sample group and in non-Asian group (p < .001). The AA genotype showed significantly lower levels of total cholesterol compared to GG + GA genotype using the recessive model with the non-Asian group (p < .05). Under the dominant model, body mass index of the GG genotype was significantly higher in Asian subgroups (p < .05). CONCLUSION PPARGC1A Gly482Ser variant impacts differently in Asian population groups.
Collapse
Affiliation(s)
- Prabhakar Bhatta
- School of Pharmacy and Life Sciences, Robert Gordon University, Aberdeen AB107GJ, UK
| | - Giovanna Bermano
- School of Pharmacy and Life Sciences, Robert Gordon University, Aberdeen AB107GJ, UK
| | - Hector C Williams
- School of Health Sciences, Robert Gordon University, Aberdeen AB107GJ, UK
| | - Rachel M Knott
- School of Pharmacy and Life Sciences, Robert Gordon University, Aberdeen AB107GJ, UK.
| |
Collapse
|
21
|
Polyphenol Effects on Cholesterol Metabolism via Bile Acid Biosynthesis, CYP7A1: A Review. Nutrients 2019; 11:nu11112588. [PMID: 31661763 PMCID: PMC6893479 DOI: 10.3390/nu11112588] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/27/2019] [Accepted: 10/09/2019] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis, the main contributor to coronary heart disease, is characterised by an accumulation of lipids such as cholesterol in the arterial wall. Reverse cholesterol transport (RCT) reduces cholesterol via its conversion into bile acids (BAs). During RCT in non-hepatic peripheral tissues, cholesterol is transferred to high-density lipoprotein (HDL) particles and returned to the liver for conversion into BAs predominantly via the rate-limiting enzyme, cholesterol 7 α-hydroxylase (CYP7A1). Numerous reports have described that polyphenol induced increases in BA excretion and corresponding reductions in total and LDL cholesterol in animal and in-vitro studies, but the process whereby this occurs has not been extensively reviewed. There are three main mechanisms by which BA excretion can be augmented: (1) increased expression of CYP7A1; (2) reduced expression of intestinal BA transporters; and (3) changes in the gut microbiota. Here we summarise the BA metabolic pathways focusing on CYP7A1, how its gene is regulated via transcription factors, diurnal rhythms, and microRNAs. Importantly, we will address the following questions: (1) Can polyphenols enhance BA secretion by modulating the CYP7A1 biosynthetic pathway? (2) Can polyphenols alter the BA pool via changes in the gut microbiota? (3) Which polyphenols are the most promising candidates for future research? We conclude that while in rodents some polyphenols induce CYP7A1 expression predominantly by the LXRα pathway, in human cells, this may occur through FXR, NF-KB, and ERK signalling. Additionally, gut microbiota is important for the de-conjugation and excretion of BAs. Puerarin, resveratrol, and quercetin are promising candidates for further research in this area.
Collapse
|
22
|
Liu X, Wang Y, Ortlund EA. First High-Resolution Crystal Structures of the Glucocorticoid Receptor Ligand-Binding Domain-Peroxisome Proliferator-Activated γ Coactivator 1- α Complex with Endogenous and Synthetic Glucocorticoids. Mol Pharmacol 2019; 96:408-417. [PMID: 31391291 PMCID: PMC6724573 DOI: 10.1124/mol.119.116806] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 07/30/2019] [Indexed: 11/22/2022] Open
Abstract
Both synthetic and endogenous glucocorticoids are important pharmaceutic drugs known to bind to the ligand-binding domain (LBD) of glucocorticoid receptor (GR), a member of the nuclear receptor (NR) superfamily. Ligand binding induces conformational changes within GR, resulting in subsequent DNA binding and differential coregulator recruitment, ultimately activating or repressing target gene expression. One of the most crucial coregulators is peroxisome proliferator-activated γ coactivator 1-α (PGC1α), which acts to regulate energy metabolism by directly interacting with GR to modulate gene expression. However, the mechanisms through which PGC1α senses GR conformation to drive transcription are not completely known. Here, an ancestral variant of the GR (AncGR2) LBD was used as a tool to produce stable protein for biochemical and structural studies. PGC1α is found to interact more tightly and form a more stable complex with AncGR2 LBD than nuclear receptor coactivator 2. We report the first high-resolution X-ray crystal structures of AncGR2 LBD in complex with PGC1α and dexamethasone (DEX) or hydrocortisone (HCY). Structural analyses reveal how distinct steroid drugs bind to GR with different affinities by unique hydrogen bonds and hydrophobic interactions. Important charge clamps are formed between the activation function 2 and PGC1α to mediate their specific interactions. These interactions lead to a high level of protection from hydrogen-deuterium exchange at the coregulator interaction site and strong intramolecular allosteric communication to ligand binding site. This is the first structure detailing the GR-PGC1α interaction providing a foundation for future design of specific therapeutic agents targeting these critical metabolic regulators. SIGNIFICANCE STATEMENT: High-resolution structures of AncGR2 LBD bound to DEX and HCY in complex with PGC1α reveal the molecular mechanism of PGC1α binding to AncGR2 LBD as well as the distinct affinities between DEX and HCY binding. Identifying the structural mechanisms that drive drug affinity is of pharmacologic interest to the glucocorticoid receptor field as an avenue to guide future drug design targeting GR-PGC1α signaling, which plays a crucial role in controlling hepatic glucose output.
Collapse
Affiliation(s)
- Xu Liu
- Department of Biochemistry, Emory University School of Medicine, Atlanta Georgia (X.L., Y.W., E.A.O.) and College of Life Sciences, Qingdao University, Qingdao, People's Republic of China (Y.W.)
| | - Yashuo Wang
- Department of Biochemistry, Emory University School of Medicine, Atlanta Georgia (X.L., Y.W., E.A.O.) and College of Life Sciences, Qingdao University, Qingdao, People's Republic of China (Y.W.)
| | - Eric A Ortlund
- Department of Biochemistry, Emory University School of Medicine, Atlanta Georgia (X.L., Y.W., E.A.O.) and College of Life Sciences, Qingdao University, Qingdao, People's Republic of China (Y.W.)
| |
Collapse
|
23
|
Activation of Mevalonate Pathway via LKB1 Is Essential for Stability of Treg Cells. Cell Rep 2019; 27:2948-2961.e7. [DOI: 10.1016/j.celrep.2019.05.020] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 04/04/2019] [Accepted: 05/02/2019] [Indexed: 12/18/2022] Open
|
24
|
Ge MX, Shao RG, He HW. Advances in understanding the regulatory mechanism of cholesterol 7α-hydroxylase. Biochem Pharmacol 2019; 164:152-164. [DOI: 10.1016/j.bcp.2019.04.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 04/08/2019] [Indexed: 02/07/2023]
|
25
|
Shumar SA, Kerr EW, Fagone P, Infante AM, Leonardi R. Overexpression of Nudt7 decreases bile acid levels and peroxisomal fatty acid oxidation in the liver. J Lipid Res 2019; 60:1005-1019. [PMID: 30846528 PMCID: PMC6495166 DOI: 10.1194/jlr.m092676] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/04/2019] [Indexed: 12/14/2022] Open
Abstract
Lipid metabolism requires CoA, an essential cofactor found in multiple subcellular compartments, including the peroxisomes. In the liver, CoA levels are dynamically adjusted between the fed and fasted states. Elevated CoA levels in the fasted state are driven by increased synthesis; however, this also correlates with decreased expression of Nudix hydrolase (Nudt)7, the major CoA-degrading enzyme in the liver. Nudt7 resides in the peroxisomes, and we overexpressed this enzyme in mouse livers to determine its effect on the size and composition of the hepatic CoA pool in the fed and fasted states. Nudt7 overexpression did not change total CoA levels, but decreased the concentration of short-chain acyl-CoAs and choloyl-CoA in fasted livers, when endogenous Nudt7 activity was lowest. The effect on these acyl-CoAs correlated with a significant decrease in the hepatic bile acid content and in the rate of peroxisomal fatty acid oxidation, as estimated by targeted and untargeted metabolomics, combined with the measurement of fatty acid oxidation in intact hepatocytes. Identification of the CoA species and metabolic pathways affected by the overexpression on Nudt7 in vivo supports the conclusion that the nutritionally driven modulation of Nudt7 activity could contribute to the regulation of the peroxisomal CoA pool and peroxisomal lipid metabolism.
Collapse
Affiliation(s)
- Stephanie A Shumar
- Department of Biochemistry, West Virginia University, Morgantown, WV 26506
| | - Evan W Kerr
- Department of Biochemistry, West Virginia University, Morgantown, WV 26506
| | - Paolo Fagone
- Department of Biochemistry, West Virginia University, Morgantown, WV 26506; Protein Core Facility West Virginia University, Morgantown, WV 26506
| | - Aniello M Infante
- Genomics Core Facility West Virginia University, Morgantown, WV 26506
| | - Roberta Leonardi
- Department of Biochemistry, West Virginia University, Morgantown, WV 26506.
| |
Collapse
|
26
|
Rennert C, Vlaic S, Marbach-Breitrück E, Thiel C, Sales S, Shevchenko A, Gebhardt R, Matz-Soja M. The Diurnal Timing of Starvation Differently Impacts Murine Hepatic Gene Expression and Lipid Metabolism - A Systems Biology Analysis Using Self-Organizing Maps. Front Physiol 2018; 9:1180. [PMID: 30271348 PMCID: PMC6146234 DOI: 10.3389/fphys.2018.01180] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 08/06/2018] [Indexed: 12/24/2022] Open
Abstract
Organisms adapt their metabolism and draw on reserves as a consequence of food deprivation. The central role of the liver in starvation response is to coordinate a sufficient energy supply for the entire organism, which has frequently been investigated. However, knowledge of how circadian rhythms impact on and alter this response is scarce. Therefore, we investigated the influence of different timings of starvation on global hepatic gene expression. Mice (n = 3 each) were challenged with 24-h food deprivation started in the morning or evening, coupled with refeeding for different lengths and compared with ad libitum fed control groups. Alterations in hepatocyte gene expression were quantified using microarrays and confirmed or complemented with qPCR, especially for lowly detectable transcription factors. Analysis was performed using self-organizing maps (SOMs), which bases on clustering genes with similar expression profiles. This provides an intuitive overview of expression trends and allows easier global comparisons between complex conditions. Transcriptome analysis revealed a strong circadian-driven response to fasting based on the diurnal expression of transcription factors (e.g., Ppara, Pparg). Starvation initiated in the morning produced known metabolic adaptations in the liver; e.g., switching from glucose storage to consumption and gluconeogenesis. However, starvation initiated in the evening produced a different expression signature that was controlled by yet unknown regulatory mechanisms. For example, the expression of genes involved in gluconeogenesis decreased and fatty acid and cholesterol synthesis genes were induced. The differential regulation after morning and evening starvation were also reflected at the lipidome level. The accumulation of hepatocellular storage lipids (triacylglycerides, cholesteryl esters) was significantly higher after the initiation of starvation in the morning compared to the evening. Concerning refeeding, the gene expression pattern after a 12 h refeeding period largely resembled that of the corresponding starvation state but approached the ad libitum control state after refeeding for 21 h. Some components of these regulatory circuits are discussed. Collectively, these data illustrate a highly time-dependent starvation response in the liver and suggest that a circadian influence cannot be neglected when starvation is the focus of research or medicine, e.g., in the case of treating victims of sudden starvation events.
Collapse
Affiliation(s)
- Christiane Rennert
- Rudolf-Schönheimer-Institute of Biochemistry, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Sebastian Vlaic
- Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany
| | - Eugenia Marbach-Breitrück
- Rudolf-Schönheimer-Institute of Biochemistry, Faculty of Medicine, Leipzig University, Leipzig, Germany.,Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Carlo Thiel
- Rudolf-Schönheimer-Institute of Biochemistry, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Susanne Sales
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Andrej Shevchenko
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Rolf Gebhardt
- Rudolf-Schönheimer-Institute of Biochemistry, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Madlen Matz-Soja
- Rudolf-Schönheimer-Institute of Biochemistry, Faculty of Medicine, Leipzig University, Leipzig, Germany
| |
Collapse
|
27
|
Supplementation with an insoluble fiber obtained from carob pod (Ceratonia siliqua L.) rich in polyphenols prevents dyslipidemia in rabbits through SIRT1/PGC-1α pathway. Eur J Nutr 2017; 58:357-366. [DOI: 10.1007/s00394-017-1599-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 12/14/2017] [Indexed: 02/07/2023]
|
28
|
p38α MAPK antagonizing JNK to control the hepatic fat accumulation in pediatric patients onset intestinal failure. Cell Death Dis 2017; 8:e3110. [PMID: 29022907 PMCID: PMC5682685 DOI: 10.1038/cddis.2017.523] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 08/23/2017] [Accepted: 09/07/2017] [Indexed: 12/30/2022]
Abstract
The p38α mitogen-activated protein kinase (MAPK) has been related to gluconeogenesis and lipid metabolism. However, the roles and related mechanisms of p38α MAPK in intestinal failure (IF)-associated liver steatosis remained poor understood. Here, our experimental evidence suggested that p38α MAPK significantly suppressed the fat accumulation in livers of IF patients mainly through two mechanisms. On the one hand, p38α MAPK increased hepatic bile acid (BA) synthesis by upregulating the expression of the rate-limiting enzyme cholesterol 7-α-hydroxylase (CYP7A1) and peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), which in turn activated the transcription of the CYP7A1. On the other hand, p38α MAPK promoted fatty acid (FA) β-oxidation via upregulating peroxisome proliferator-activated receptor alpha (PPARα) and its transcriptional target genes carnitine palmitoyltransferase 1A (CPT1A) and peroxisomal acyl-coenzyme aoxidase 1 (ACOX1). Dual luciferase assays indicated that p38α MAPK increased the transcription of PPARα, PGC-1α and CYP7A1 by upregulating their promoters’ activities. In addition, in vitro and in vivo assays indicated p38α MAPK negatively regulates the hepatic steatosis by controlling JNK activation. In conculsion, our findings demonstrate that hepatic p38α MAPK functions as a negative regulator of liver steatosis in maintaining BA synthesis and FAO by antagonizing the c-Jun N-terminal kinase (JNK).
Collapse
|
29
|
Golubeva AV, Joyce SA, Moloney G, Burokas A, Sherwin E, Arboleya S, Flynn I, Khochanskiy D, Moya-Pérez A, Peterson V, Rea K, Murphy K, Makarova O, Buravkov S, Hyland NP, Stanton C, Clarke G, Gahan CGM, Dinan TG, Cryan JF. Microbiota-related Changes in Bile Acid & Tryptophan Metabolism are Associated with Gastrointestinal Dysfunction in a Mouse Model of Autism. EBioMedicine 2017; 24:166-178. [PMID: 28965876 PMCID: PMC5652137 DOI: 10.1016/j.ebiom.2017.09.020] [Citation(s) in RCA: 224] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 09/01/2017] [Accepted: 09/15/2017] [Indexed: 01/24/2023] Open
Abstract
Autism spectrum disorder (ASD) is one of the most prevalent neurodevelopmental conditions worldwide. There is growing awareness that ASD is highly comorbid with gastrointestinal distress and altered intestinal microbiome, and that host-microbiome interactions may contribute to the disease symptoms. However, the paucity of knowledge on gut-brain axis signaling in autism constitutes an obstacle to the development of precision microbiota-based therapeutics in ASD. To this end, we explored the interactions between intestinal microbiota, gut physiology and social behavior in a BTBR T+Itpr3tf/J mouse model of ASD. Here we show that a reduction in the relative abundance of very particular bacterial taxa in the BTBR gut - namely, bile-metabolizing Bifidobacterium and Blautia species, - is associated with deficient bile acid and tryptophan metabolism in the intestine, marked gastrointestinal dysfunction, as well as impaired social interactions in BTBR mice. Together these data support the concept of targeted manipulation of the gut microbiota for reversing gastrointestinal and behavioral symptomatology in ASD, and offer specific plausible targets in this endeavor.
Collapse
Affiliation(s)
- Anna V Golubeva
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Susan A Joyce
- APC Microbiome Institute, University College Cork, Cork, Ireland; School of Biochemistry & Cell Biology, University College Cork, Cork, Ireland
| | - Gerard Moloney
- Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland
| | | | - Eoin Sherwin
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Silvia Arboleya
- APC Microbiome Institute, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark Fermoy, County Cork, Ireland
| | - Ian Flynn
- Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland
| | | | | | | | - Kieran Rea
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Kiera Murphy
- Teagasc Food Research Centre, Moorepark Fermoy, County Cork, Ireland
| | - Olga Makarova
- Research Institute of Human Morphology, Moscow, Russia; Lomonosov Moscow State University, Moscow, Russia
| | - Sergey Buravkov
- Research Institute of Human Morphology, Moscow, Russia; Lomonosov Moscow State University, Moscow, Russia
| | - Niall P Hyland
- APC Microbiome Institute, University College Cork, Cork, Ireland; Department of Pharmacology & Therapeutics, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Institute, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark Fermoy, County Cork, Ireland; Department of Psychiatry & Neurobehavioural Sciences, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Institute, University College Cork, Cork, Ireland; Department of Psychiatry & Neurobehavioural Sciences, University College Cork, Cork, Ireland
| | - Cormac G M Gahan
- APC Microbiome Institute, University College Cork, Cork, Ireland; School of Microbiology, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Institute, University College Cork, Cork, Ireland; Department of Psychiatry & Neurobehavioural Sciences, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Institute, University College Cork, Cork, Ireland; Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
30
|
Tobina T, Mori Y, Doi Y, Nakayama F, Kiyonaga A, Tanaka H. Peroxisome proliferator-activated receptor gamma co-activator 1 gene Gly482Ser polymorphism is associated with the response of low-density lipoprotein cholesterol concentrations to exercise training in elderly Japanese. J Physiol Sci 2017; 67:595-602. [PMID: 27699582 PMCID: PMC10717479 DOI: 10.1007/s12576-016-0491-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/20/2016] [Indexed: 01/20/2023]
Abstract
Muscle peroxisome proliferator-activated receptor gamma co-activator 1 (PGC-1)α gene expression is influenced by the Gly482Ser gene polymorphism, which is a candidate genetic risk factor for diabetes mellitus and obesity. This study investigated the effects of PGC-1 gene Gly482Ser polymorphisms on alterations in glucose and lipid metabolism induced by exercise training. A 12-week intervention study was performed for 119 participants who were more than 65 years of age and completed exercise training at lactate threshold intensity. Total cholesterol and low-density lipoprotein cholesterol were significantly reduced in Gly/Gly but not in Gly/Ser and Ser/Ser participants after exercise. The Gly/Gly genotype of the PGC-1 gene Gly482Ser polymorphism influences the effects of moderate-intensity exercise training on low-density lipoprotein cholesterol and total cholesterol concentrations in older people.
Collapse
Affiliation(s)
- Takuro Tobina
- Faculty of Nursing and Nutrition, University of Nagasaki, Manabino1-1-1, Nagayo-cho, Nishisonogi-gun, Nagasaki, 851-2195, Japan.
- The Fukuoka University Institute for Physical Activity, Fukuoka University, Fukuoka, Japan.
| | - Yukari Mori
- Faculty of Sports Science, Fukuoka University, Fukuoka, Japan
| | - Yukiko Doi
- The Fukuoka University Institute for Physical Activity, Fukuoka University, Fukuoka, Japan
- Faculty of Commerce, Department of Tourism Industry, Kyushu Sangyo University, Fukuoka, Japan
| | - Fuki Nakayama
- Faculty of Sports Science, Fukuoka University, Fukuoka, Japan
| | - Akira Kiyonaga
- The Fukuoka University Institute for Physical Activity, Fukuoka University, Fukuoka, Japan
- Faculty of Sports Science, Fukuoka University, Fukuoka, Japan
| | - Hiroaki Tanaka
- The Fukuoka University Institute for Physical Activity, Fukuoka University, Fukuoka, Japan
- Faculty of Sports Science, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
31
|
Zhang Y, Kim DK, Lee JM, Park SB, Jeong WI, Kim SH, Lee IK, Lee CH, Chiang JYL, Choi HS. Orphan nuclear receptor oestrogen-related receptor γ (ERRγ) plays a key role in hepatic cannabinoid receptor type 1-mediated induction of CYP7A1 gene expression. Biochem J 2015; 470:181-93. [PMID: 26348907 PMCID: PMC5333639 DOI: 10.1042/bj20141494] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 06/29/2015] [Indexed: 12/30/2022]
Abstract
Bile acids are primarily synthesized from cholesterol in the liver and have important roles in dietary lipid absorption and cholesterol homoeostasis. Detailed roles of the orphan nuclear receptors regulating cholesterol 7α-hydroxylase (CYP7A1), the rate-limiting enzyme in bile acid synthesis, have not yet been fully elucidated. In the present study, we report that oestrogen-related receptor γ (ERRγ) is a novel transcriptional regulator of CYP7A1 expression. Activation of cannabinoid receptor type 1 (CB1 receptor) signalling induced ERRγ-mediated transcription of the CYP7A1 gene. Overexpression of ERRγ increased CYP7A1 expression in vitro and in vivo, whereas knockdown of ERRγ attenuated CYP7A1 expression. Deletion analysis of the CYP7A1 gene promoter and a ChIP assay revealed an ERRγ-binding site on the CYP7A1 gene promoter. Small heterodimer partner (SHP) inhibited the transcriptional activity of ERRγ and thus regulated CYP7A1 expression. Overexpression of ERRγ led to increased bile acid levels, whereas an inverse agonist of ERRγ, GSK5182, reduced CYP7A1 expression and bile acid synthesis. Finally, GSK5182 significantly reduced hepatic CB1 receptor-mediated induction of CYP7A1 expression and bile acid synthesis in alcohol-treated mice. These results provide the molecular mechanism linking ERRγ and bile acid metabolism.
Collapse
MESH Headings
- Animals
- Bile Acids and Salts/metabolism
- Cells, Cultured
- Cholesterol 7-alpha-Hydroxylase/biosynthesis
- Cholesterol 7-alpha-Hydroxylase/genetics
- Drug Inverse Agonism
- Ethanol/pharmacology
- Gene Expression
- Glycerides/pharmacology
- HEK293 Cells
- Hepatocytes/metabolism
- Humans
- Liver/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Promoter Regions, Genetic
- Rats, Sprague-Dawley
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/genetics
- Receptor, Cannabinoid, CB1/metabolism
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Transcription, Genetic
Collapse
Affiliation(s)
- Yaochen Zhang
- National Creative Research Initiatives Center for Nuclear Receptor Signals and Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Don-Kyu Kim
- National Creative Research Initiatives Center for Nuclear Receptor Signals and Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Ji-Min Lee
- National Creative Research Initiatives Center for Nuclear Receptor Signals and Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Seung Bum Park
- Chemical Biology Laboratory, School of Chemistry, Seoul National University, Seoul 151-742, Republic of Korea
| | - Won-Il Jeong
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 305-338, Republic of Korea
| | - Seong Heon Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 701-310, Republic of Korea
| | - In-Kyu Lee
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu 700-721, Republic of Korea
| | - Chul-Ho Lee
- Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea
| | - John Y L Chiang
- Department of Integrative Medical Sciences, Northeastern Ohio University's Colleges of Medicine and Pharmacy, Rootstown, Ohio 44272, U.S.A
| | - Hueng-Sik Choi
- National Creative Research Initiatives Center for Nuclear Receptor Signals and Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Republic of Korea
| |
Collapse
|
32
|
Chen Y, Wang Z, Xu M, Wang X, Liu R, Liu Q, Zhang Z, Xia T, Zhao J, Jiang G, Xu Y, Liu S. Nanosilver incurs an adaptive shunt of energy metabolism mode to glycolysis in tumor and nontumor cells. ACS NANO 2014; 8:5813-5825. [PMID: 24810997 DOI: 10.1021/nn500719m] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Due to its significant antimicrobial properties, nanosilver (nAg) has been substantially used in a wide spectrum of areas. This has raised the concerns on the detrimental effects on environment and human health. Although numerous studies have documented nAg-mediated toxicity to cells or organisms, little attempt has been made to study the biological impacts of nAg on cells at nontoxic concentrations, namely, the distinct biological effects that can be separated from direct cytotoxicity. Here, we studied nAg-mediated effects on energy metabolism in cells under sublethal exposure. Treatment of nAg at nontoxic concentrations resulted in a decline of ATP synthesis and attenuation of respiratory chain function in nontumor HEK293T cells and tumor cells with differential respiration rate, including HepG2, HeLa, A498, and PC3 cells. Cellular energy homeostasis was switched from oxidative phosphorylation-based aerobic metabolism to anaerobic glycolysis, which is an adaption process to satisfy the energy demand for cell survival. Nanospheres with smaller size showed greater capability to alter cellular energy metabolism than those with larger size or nanoplates. Mechanistic investigation manifested that inhibition of PGC-1α by nAg was, at least partially, accountable for the transition from oxidative phosphorylation to glycolysis. Additionally, altered expression of a few energy metabolism-related genes (such as PFKFB3 and PDHA1) was also involved in the transition process. We further showed nAg-induced depolarization of mitochondrial membrane potential and reduction of respiratory chain complex activity. Together, our combined results uncovered the mechanisms by which nAg induced energy metabolism reprogramming in both tumor and nontumor cells under sublethal dosage.
Collapse
Affiliation(s)
- Yue Chen
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences , Beijing 100085, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Functional crosstalk of PGC-1 coactivators and inflammation in skeletal muscle pathophysiology. Semin Immunopathol 2013; 36:27-53. [DOI: 10.1007/s00281-013-0406-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 10/29/2013] [Indexed: 02/06/2023]
|
34
|
Abstract
Bile acids are important physiological agents for intestinal nutrient absorption and biliary secretion of lipids, toxic metabolites, and xenobiotics. Bile acids also are signaling molecules and metabolic regulators that activate nuclear receptors and G protein-coupled receptor (GPCR) signaling to regulate hepatic lipid, glucose, and energy homeostasis and maintain metabolic homeostasis. Conversion of cholesterol to bile acids is critical for maintaining cholesterol homeostasis and preventing accumulation of cholesterol, triglycerides, and toxic metabolites, and injury in the liver and other organs. Enterohepatic circulation of bile acids from the liver to intestine and back to the liver plays a central role in nutrient absorption and distribution, and metabolic regulation and homeostasis. This physiological process is regulated by a complex membrane transport system in the liver and intestine regulated by nuclear receptors. Toxic bile acids may cause inflammation, apoptosis, and cell death. On the other hand, bile acid-activated nuclear and GPCR signaling protects against inflammation in liver, intestine, and macrophages. Disorders in bile acid metabolism cause cholestatic liver diseases, dyslipidemia, fatty liver diseases, cardiovascular diseases, and diabetes. Bile acids, bile acid derivatives, and bile acid sequestrants are therapeutic agents for treating chronic liver diseases, obesity, and diabetes in humans.
Collapse
|
35
|
Fu ZD, Klaassen CD. Increased bile acids in enterohepatic circulation by short-term calorie restriction in male mice. Toxicol Appl Pharmacol 2013; 273:680-90. [PMID: 24183703 DOI: 10.1016/j.taap.2013.10.020] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 10/16/2013] [Accepted: 10/20/2013] [Indexed: 12/15/2022]
Abstract
Previous studies showed glucose and insulin signaling can regulate bile acid (BA) metabolism during fasting or feeding. However, limited knowledge is available on the effect of calorie restriction (CR), a well-known anti-aging intervention, on BA homeostasis. To address this, the present study utilized a "dose-response" model of CR, where male C57BL/6 mice were fed 0, 15, 30, or 40% CR diets for one month, followed by BA profiling in various compartments of the enterohepatic circulation by UPLC-MS/MS technique. This study showed that 40% CR increased the BA pool size (162%) as well as total BAs in serum, gallbladder, and small intestinal contents. In addition, CR "dose-dependently" increased the concentrations of tauro-cholic acid (TCA) and many secondary BAs (produced by intestinal bacteria) in serum, such as tauro-deoxycholic acid (TDCA), DCA, lithocholic acid, ω-muricholic acid (ωMCA), and hyodeoxycholic acid. Notably, 40% CR increased TDCA by over 1000% (serum, liver, and gallbladder). Interestingly, 40% CR increased the proportion of 12α-hydroxylated BAs (CA and DCA), which correlated with improved glucose tolerance and lipid parameters. The CR-induced increase in BAs correlated with increased expression of BA-synthetic (Cyp7a1) and conjugating enzymes (BAL), and the ileal BA-binding protein (Ibabp). These results suggest that CR increases BAs in male mice possibly through orchestrated increases in BA synthesis and conjugation in liver as well as intracellular transport in ileum.
Collapse
Affiliation(s)
- Zidong Donna Fu
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | | |
Collapse
|
36
|
McCarthy C, Lieggi NT, Barry D, Mooney D, de Gaetano M, James WG, McClelland S, Barry MC, Escoubet-Lozach L, Li AC, Glass CK, Fitzgerald DJ, Belton O. Macrophage PPAR gamma Co-activator-1 alpha participates in repressing foam cell formation and atherosclerosis in response to conjugated linoleic acid. EMBO Mol Med 2013; 5:1443-57. [PMID: 23964012 PMCID: PMC3799497 DOI: 10.1002/emmm.201302587] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 06/27/2013] [Accepted: 07/02/2013] [Indexed: 12/31/2022] Open
Abstract
Conjugated linoleic acid (CLA) has the unique property of inducing regression of pre-established murine atherosclerosis. Understanding the mechanism(s) involved may help identify endogenous pathways that reverse human atherosclerosis. Here, we provide evidence that CLA inhibits foam cell formation via regulation of the nuclear receptor coactivator, peroxisome proliferator-activated receptor (PPAR)-γ coactivator (PGC)-1α, and that macrophage PGC-1α plays a role in atheroprotection in vivo. PGC-1α was identified as a hub gene within a cluster in the aorta of the apoE−/− mouse in the CLA-induced regression model. PGC-1α was localized to macrophage/foam cells in the murine aorta where its expression was increased during CLA-induced regression. PGC-1α expression was also detected in macrophages in human atherosclerosis and was inversely linked to disease progression in patients with the disease. Deletion of PGC-1α in bone marrow derived macrophages promoted, whilst over expression of the gene inhibited foam cell formation. Importantly, macrophage specific deletion of PGC-1α accelerated atherosclerosis in the LDLR−/− mouse in vivo. These novel data support a functional role for PGC-1α in atheroprotection.
Collapse
Affiliation(s)
- Cathal McCarthy
- School of Biomolecular and Biomedical Science, UCD Conway Institute, UCD, Dublin, Ireland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Wegner CJ, Kim B, Lee J. Trust your gut: galvanizing nutritional interest in intestinal cholesterol metabolism for protection against cardiovascular diseases. Nutrients 2013; 5:208-22. [PMID: 23325147 PMCID: PMC3571644 DOI: 10.3390/nu5010208] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 01/09/2013] [Accepted: 01/11/2013] [Indexed: 02/08/2023] Open
Abstract
Recent studies have demonstrated that the intestine is a key target organ for overall health and longevity. Complementing these studies is the discovery of the trans-intestinal cholesterol efflux pathway and the emerging role of the intestine in reverse cholesterol transport. The surfacing dynamics of the regulation of cholesterol metabolism in the intestine provides an attractive platform for intestine-specific nutritional intervention strategies to lower blood cholesterol levels for protection against cardiovascular diseases. Notably, there is mounting evidence that stimulation of pathways associated with calorie restriction may have a large effect on the regulation of cholesterol removal by the intestine. However, intestinal energy metabolism, specifically the idiosyncrasies surrounding intestinal responses to energy deprivation, is poorly understood. The goal of this paper is to review recent insights into cholesterol regulation by the intestine and to discuss the potential for positive regulation of intestine-driven cholesterol removal through the nutritional induction of pathways associated with calorie restriction.
Collapse
Affiliation(s)
- Casey J Wegner
- Department of Nutritional Sciences, University of Connecticut, 216 Advanced Technology Laboratory Building, 1392 Storrs Road, Storrs, CT 06269, USA.
| | | | | |
Collapse
|
38
|
Raymond F, Wang L, Moser M, Metairon S, Mansourian R, Zwahlen MC, Kussmann M, Fuerholz A, Macé K, Chou CJ. Consequences of exchanging carbohydrates for proteins in the cholesterol metabolism of mice fed a high-fat diet. PLoS One 2012; 7:e49058. [PMID: 23139832 PMCID: PMC3490911 DOI: 10.1371/journal.pone.0049058] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 10/09/2012] [Indexed: 01/25/2023] Open
Abstract
Consumption of low-carbohydrate, high-protein, high-fat diets lead to rapid weight loss but the cardioprotective effects of these diets have been questioned. We examined the impact of high-protein and high-fat diets on cholesterol metabolism by comparing the plasma cholesterol and the expression of cholesterol biosynthesis genes in the liver of mice fed a high-fat (HF) diet that has a high (H) or a low (L) protein-to-carbohydrate (P/C) ratio. H-P/C-HF feeding, compared with L-P/C-HF feeding, decreased plasma total cholesterol and increased HDL cholesterol concentrations at 4-wk. Interestingly, the expression of genes involved in hepatic steroid biosynthesis responded to an increased dietary P/C ratio by first down-regulation (2-d) followed by later up-regulation at 4-wk, and the temporal gene expression patterns were connected to the putative activity of SREBF1 and 2. In contrast, Cyp7a1, the gene responsible for the conversion of cholesterol to bile acids, was consistently up-regulated in the H-P/C-HF liver regardless of feeding duration. Over expression of Cyp7a1 after 2-d and 4-wk H-P/C-HF feeding was connected to two unique sets of transcription regulators. At both time points, up-regulation of the Cyp7a1 gene could be explained by enhanced activations and reduced suppressions of multiple transcription regulators. In conclusion, we demonstrated that the hypocholesterolemic effect of H-P/C-HF feeding coincided with orchestrated changes of gene expressions in lipid metabolic pathways in the liver of mice. Based on these results, we hypothesize that the cholesterol lowering effect of high-protein feeding is associated with enhanced bile acid production but clinical validation is warranted. (246 words).
Collapse
Affiliation(s)
- Frédéric Raymond
- Bioanalytical Science Department, Nestlé Research Center, Lausanne, Switzerland
| | - Long Wang
- Department of Nutrition Science and Dietetics, Syracuse University, Syracuse, New York, United States of America
| | - Mireille Moser
- Bioanalytical Science Department, Nestlé Research Center, Lausanne, Switzerland
| | - Sylviane Metairon
- Bioanalytical Science Department, Nestlé Research Center, Lausanne, Switzerland
| | - Robert Mansourian
- Bioanalytical Science Department, Nestlé Research Center, Lausanne, Switzerland
| | | | - Martin Kussmann
- Proteomics and Metabonomics Core, Nestlé Institute of Health Sciences, Lausanne, Switzerland
- Faculty of Science, Aarhus University, Aarhus, Denmark
- Faculty of Life Sciences, Federal Institute of Technology, Lausanne, Switzerland
| | - Andreas Fuerholz
- Bioanalytical Science Department, Nestlé Research Center, Lausanne, Switzerland
| | - Katherine Macé
- Nutrition and Health Department, Nestlé Research Center, Lausanne, Switzerland
| | - Chieh Jason Chou
- Nutrition and Health Department, Nestlé Research Center, Lausanne, Switzerland
| |
Collapse
|
39
|
Novotna A, Doricakova A, Pavek P, Dvorak Z. Construction and characterization of peroxisome proliferator-activated receptor-gamma co-activator 1 alpha (PGC-1α over-expressing cell line derived from human hepatocyte carcinoma HepG2 cells). Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2012; 157:214-21. [PMID: 23073537 DOI: 10.5507/bp.2012.075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 07/13/2012] [Indexed: 11/23/2022] Open
Abstract
AIMS The aim was develop stable human cell line stable over-expressing transcription co-activator peroxisome proliferator-activated receptor gamma co-activator 1α (PGC-1α) with restored hepatospecific functions and increased expression of major xenobiotic metabolizing enzymes. METHODS Six clones of HepG2-PGC-1α and one control clone HepG2-pcDNA3 were isolated and analyzed for secretion of hepatospecific markers, fibrinogen, albumin and alpha1-antitrypsin. Expression levels of protein and mRNA of hepatocyte nuclear factor (HNF4α), pregnane X receptor (PXR) and aryl hydrocarbon receptor (AhR) were determined. We measured basal and ligand inducible expression of CYP1A1 and CYP3A4. RESULTS Stably transfected cell line HepG2-PGC-1α derived from HepG2 cells over-expressing PGC-1α displayed increased secretion of fibrinogen, but not albumin or alpha1-antitrypsin compared to parent HepG2 cells. We found increased levels of HNF4α, PXR and AhR proteins but not their mRNAs in HepG2-PGC1 cells. Basal expression of CYP3A4 protein in HepG2-PGC-1α cells was increased but rifampicin-inducible expression of CYP3A4 protein was lowered in comparison with parent HepG2 cells. Induction of CYP3A4 mRNA varied between 1.3 - 1.9 fold in individual clones. Expression of TCDD-inducible CYP1A1 protein was lower in HepG2-PGC-1α cells than in parent HepG2 cells. Induction of CYP1A1 mRNA by TCDD in HepG2-PGC-1α cells was comparable with that in parent HepG2 cells and ranged between 103 - 198 fold. CONCLUSION Stable expression of PGC-1α in HepG2 cells restores several hepatospecific functions, such as secretion of fibrinogen, expression of HNF4α1 and xenoreceptors PXR and AhR. However, the expression and induction of key drug-metabolizing enzymes (CYP1A1 and CYP3A4) were not improved.
Collapse
Affiliation(s)
- Aneta Novotna
- Department of Cell Biology and Genetics, Faculty of Science, Palacky University Olomouc, Czech Republic
| | | | | | | |
Collapse
|
40
|
Cantó C, Houtkooper RH, Pirinen E, Youn DY, Oosterveer MH, Cen Y, Fernandez-Marcos PJ, Yamamoto H, Andreux PA, Cettour-Rose P, Gademann K, Rinsch C, Schoonjans K, Sauve AA, Auwerx J. The NAD(+) precursor nicotinamide riboside enhances oxidative metabolism and protects against high-fat diet-induced obesity. Cell Metab 2012; 15:838-47. [PMID: 22682224 PMCID: PMC3616313 DOI: 10.1016/j.cmet.2012.04.022] [Citation(s) in RCA: 885] [Impact Index Per Article: 73.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 03/02/2012] [Accepted: 04/25/2012] [Indexed: 01/05/2023]
Abstract
As NAD(+) is a rate-limiting cosubstrate for the sirtuin enzymes, its modulation is emerging as a valuable tool to regulate sirtuin function and, consequently, oxidative metabolism. In line with this premise, decreased activity of PARP-1 or CD38-both NAD(+) consumers-increases NAD(+) bioavailability, resulting in SIRT1 activation and protection against metabolic disease. Here we evaluated whether similar effects could be achieved by increasing the supply of nicotinamide riboside (NR), a recently described natural NAD(+) precursor with the ability to increase NAD(+) levels, Sir2-dependent gene silencing, and replicative life span in yeast. We show that NR supplementation in mammalian cells and mouse tissues increases NAD(+) levels and activates SIRT1 and SIRT3, culminating in enhanced oxidative metabolism and protection against high-fat diet-induced metabolic abnormalities. Consequently, our results indicate that the natural vitamin NR could be used as a nutritional supplement to ameliorate metabolic and age-related disorders characterized by defective mitochondrial function.
Collapse
Affiliation(s)
- Carles Cantó
- École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Park WH, Pak YK. Insulin-dependent suppression of cholesterol 7α-hydroxylase is a possible link between glucose and cholesterol metabolisms. Exp Mol Med 2012; 43:571-9. [PMID: 21817852 DOI: 10.3858/emm.2011.43.10.064] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Cholesterol 7α-hydroxylase (CYP7A1) regulates the balance between cholesterol supply and metabolism by catalyzing the rate-limiting step of bile acid biosynthesis. The transcriptional activity of CYP7A1 is tightly controlled by various nuclear receptors. A forkhead transcription factor O1 (FOXO1) plays a critical role in metabolism, and insulin inactivates FOXO1 through Akt-dependent phosphorylation and nuclear exclusion. We investigated the role of insulin- Akt-FOXO1 signaling pathway in CYP7A1 transcriptional regulation since we found putative insulin-response elements, FOXO1 binding sequences, in both rat and human CYP7A1 promoters. However, ectopic expression of FOXO1 increased the rat CYP7A1-, but mildly reduced human CYP7A1-promoter activities in a dose-dependent manner. Similarly to bile acids, insulin treatment increased small heterodimer partner (SHP) mRNA rapidly and transiently, leading to the suppression of CYP7A1 transcription in both human and rodents. Chromatin immunoprecipitation showed that FOXO1 directly bound to rat CYP1A1 promoter in the absence of insulin. FOXO1 binding to the rat promoter was diminished by insulin treatment as well as by expression of SHP. Our results suggest that the stimulation of insulin- signaling pathway of Akt-FOXO1 and SHP expression may regulate cholesterol/bile acid metabolisms in liver, linking carbohydrate and cholesterol metabolic pathways. A prolonged exposure of insulin in hyperinsulinemic insulin resistance or diabetic status represses CYP7A1 transcription and bile acid biosynthesis through SHP induction and FOXO1 inactivation, leading to impairment of the hepatic cholesterol/bile acid metabolisms.
Collapse
Affiliation(s)
- Wook Ha Park
- Department of Physiology, Department of Neuroscience College of Medicine, Kyung Hee University, Seoul, Korea
| | | |
Collapse
|
42
|
Fernández-Barrena MG, Monte MJ, Latasa MU, Uriarte I, Vicente E, Chang HCY, Rodriguez-Ortigosa CM, Elferink RO, Berasain C, Marin JJG, Prieto J, Ávila MA. Lack of Abcc3 expression impairs bile-acid induced liver growth and delays hepatic regeneration after partial hepatectomy in mice. J Hepatol 2012; 56:367-73. [PMID: 21756856 DOI: 10.1016/j.jhep.2011.05.031] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 05/10/2011] [Accepted: 05/23/2011] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS Bile acids (BA) are increasingly recognized as important modulators of liver regeneration. Increased enterohepatic BA flux has been proposed to generate specific signals that activate hepatocyte proliferation after partial hepatectomy (PH). We have investigated the role of the BA membrane transporter Mrp3 (Abcc3), which is expressed in the liver and gut, in the hepatic growth response elicited by BA and in liver regeneration after PH. METHODS Liver growth and regeneration, and the expression of growth-related genes, were studied in Mrp3(+/+) and Mrp3(-/-) mice fed a cholic acid (CA) supplemented diet and after 2/3 PH. Activation of the BA receptor FXR was measured in mice after in vivo transduction of the liver with a FXR-Luciferase reporter plasmid. BA levels were measured in portal serum and liver tissue by high performance liquid chromatography-tandem mass spectrometry. RESULTS Liver growth elicited by CA feeding was significantly reduced in Mrp3(-/-) mice. These animals showed reduced FXR activation in the liver after CA administration and decreased portal serum levels of BA. Liver regeneration after PH was significantly delayed in Mrp3-deficient mice. Proliferation-related gene expression and peak DNA synthesis in Mrp3(-/-) mice occurred later than in wild types, coinciding with a retarded elevation in intra-hepatic BA levels. CONCLUSIONS Lack of Abcc3 expression markedly impairs liver growth in response to BA and after PH. Our data suggest that Mrp3 plays a non-redundant role in the regulation of BA flux during liver regeneration.
Collapse
|
43
|
Zhang Z, Liu J, Xi Y, Yang R, Chen H, Li Z, Liu D, Liang C. Two novel cis-elements involved in hepatocyte nuclear factor 4α regulation of acyl-coenzyme A:cholesterol acyltransferase 2 expression. Acta Biochim Biophys Sin (Shanghai) 2012; 44:162-71. [PMID: 22155889 DOI: 10.1093/abbs/gmr102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Acyl-coenzyme A:cholesterol acyltransferase 2 (ACAT2) is important for cholesterol ester synthesis and secretion. A previous study revealed that ACAT2 gene promoter activity was upregulated by hepatocyte nuclear factor 4α (HNF4α) through two sites around -247 and -311 of ACAT2 gene promoter. Here, we identified two novel cis-elements, site I (-1006 to -898) and site II (-38 to -29), which are important for HNF4α effect. In HepG2 cells, mutation of site I decreased ACAT2 gene promoter activity to one-fifth of that of the wild type, while mutation of site II reduced promoter activity to less than one-tenth of that of the wild type. In 293T cells, mutation of these two cis-elements profoundly impaired the HNF4α induction effect. When either of these two elements was inserted into pGL3-promoter, HNF4α induced promoter activity through the inserted element, while mutation of the element impaired HNF4α induction effect. In electrophoretic mobility shift assay and chromatin immunoprecipitation experiment, HNF4α bound to these two elements. Thus, the two cis-elements are important for HNF4α effect on ACAT2 gene transcription. We also showed that HNF4α positively regulates ACAT2 gene expression at mRNA level. Overexpression of HNF4α increased ACAT2 expression, whereas knockdown of HNF4α decreased ACAT2 expression. Peroxisome proliferator-activated receptor gamma coactivator 1α (PCG1α), a coactivator of HNF4α, increased ACAT2 expression, while small heterodimer partner (SHP), a corepressor of HNF4α, decreased ACAT2 expression. These results provide more insights into transcriptional regulation of ACAT2 expression.
Collapse
Affiliation(s)
- Zhuqin Zhang
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Science, Chinese Academy of Medical Sciences, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Li T, Francl JM, Boehme S, Ochoa A, Zhang Y, Klaassen CD, Erickson SK, Chiang JYL. Glucose and insulin induction of bile acid synthesis: mechanisms and implication in diabetes and obesity. J Biol Chem 2011; 287:1861-73. [PMID: 22144677 DOI: 10.1074/jbc.m111.305789] [Citation(s) in RCA: 159] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Bile acids facilitate postprandial absorption of nutrients. Bile acids also activate the farnesoid X receptor (FXR) and the G protein-coupled receptor TGR5 and play a major role in regulating lipid, glucose, and energy metabolism. Transgenic expression of cholesterol 7α-hydroxylase (CYP7A1) prevented high fat diet-induced diabetes and obesity in mice. In this study, we investigated the nutrient effects on bile acid synthesis. Refeeding of a chow diet to fasted mice increased CYP7A1 expression, bile acid pool size, and serum bile acids in wild type and humanized CYP7A1-transgenic mice. Chromatin immunoprecipitation assays showed that glucose increased histone acetylation and decreased histone methylation on the CYP7A1 gene promoter. Refeeding also induced CYP7A1 in fxr-deficient mice, indicating that FXR signaling did not play a role in postprandial regulation of bile acid synthesis. In streptozocin-induced type I diabetic mice and genetically obese type II diabetic ob/ob mice, hyperglycemia increased histone acetylation status on the CYP7A1 gene promoter, leading to elevated basal Cyp7a1 expression and an enlarged bile acid pool with altered bile acid composition. However, refeeding did not further increase CYP7A1 expression in diabetic mice. In summary, this study demonstrates that glucose and insulin are major postprandial factors that induce CYP7A1 gene expression and bile acid synthesis. Glucose induces CYP7A1 gene expression mainly by epigenetic mechanisms. In diabetic mice, CYP7A1 chromatin is hyperacetylated, and fasting to refeeding response is impaired and may exacerbate metabolic disorders in diabetes.
Collapse
Affiliation(s)
- Tiangang Li
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio 44272, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Teng CT, Li Y, Stockton P, Foley J. Fasting induces the expression of PGC-1α and ERR isoforms in the outer stripe of the outer medulla (OSOM) of the mouse kidney. PLoS One 2011; 6:e26961. [PMID: 22073226 PMCID: PMC3208565 DOI: 10.1371/journal.pone.0026961] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Accepted: 10/07/2011] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Peroxisome proliferator-activated receptor-γ co-activator-1α (PGC-1α) is a member of the transcriptional coactivator family that plays a central role in the regulation of cellular energy metabolism under various physiological stimuli. During fasting, PGC-1α is induced in the liver and together with estrogen-related receptor a and γ (ERRα and ERRγ, orphan nuclear receptors with no known endogenous ligand, regulate sets of genes that participate in the energy balance program. We found that PGC-1α, ERRα and ERRγ was highly expressed in human kidney HK2 cells and that PGC-1α induced dynamic protein interactions on the ERRα chromatin. However, the effect of fasting on the expression of endogenous PGC-1α, ERRα and ERRγ in the kidney is not known. METHODOLOGY/PRINCIPAL FINDINGS In this study, we demonstrated by qPCR that the expression of PGC-1α, ERRα and ERRγ was increased in the mouse kidney after fasting. By using immunohistochemistry (IHC), we showed these three proteins are co-localized in the outer stripe of the outer medulla (OSOM) of the mouse kidney. We were able to collect this region from the kidney using the Laser Capture Microdissection (LCM) technique. The qPCR data showed significant increase of PGC-1α, ERRα and ERRγ mRNA in the LCM samples after fasting for 24 hours. Furthermore, the known ERRα target genes, mitochondrial oxidative phosphorylation gene COX8H and the tricarboxylic acid (TCA) cycle gene IDH3A also showed an increase. Taken together, our data suggest that fasting activates the energy balance program in the OSOM of the kidney.
Collapse
Affiliation(s)
- Christina T Teng
- Biomolecular Screening Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, United States of America.
| | | | | | | |
Collapse
|
46
|
Bile Acid signaling in liver metabolism and diseases. J Lipids 2011; 2012:754067. [PMID: 21991404 PMCID: PMC3185234 DOI: 10.1155/2012/754067] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Accepted: 08/04/2011] [Indexed: 12/12/2022] Open
Abstract
Obesity, diabetes, and metabolic syndromes are increasingly recognized as health concerns worldwide. Overnutrition and insulin resistance are the major causes of diabetic hyperglycemia and hyperlipidemia in humans. Studies in the past decade provide evidence that bile acids are not just biological detergents facilitating gut nutrient absorption, but also important metabolic regulators of glucose and lipid homeostasis. Pharmacological alteration of bile acid metabolism or bile acid signaling pathways such as using bile acid receptor agonists or bile acid binding resins may be a promising therapeutic strategy for the treatment of obesity and diabetes. On the other hand, bile acid signaling is complex, and the molecular mechanisms mediating the bile acid effects are still not completely understood. This paper will summarize recent advances in our understanding of bile acid signaling in regulation of glucose and lipid metabolism, and the potentials of developing novel therapeutic strategies that target bile acid metabolism for the treatment of metabolic disorders.
Collapse
|
47
|
Sánchez-Ramos C, Tierrez A, Fabregat-Andrés O, Wild B, Sánchez-Cabo F, Arduini A, Dopazo A, Monsalve M. PGC-1α regulates translocated in liposarcoma activity: role in oxidative stress gene expression. Antioxid Redox Signal 2011; 15:325-37. [PMID: 21338289 DOI: 10.1089/ars.2010.3643] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
UNLABELLED Translocated in liposarcoma (TLS) is a poorly characterized multifunctional protein involved in the genotoxic response. TLS regulates gene expression at several steps, including splicing and mRNA transport, possibly connecting transcriptional and posttranscriptional events. AIMS In this study we aimed to idenfity molecular targets and regulatory partners of TLS. RESULTS AND INNOVATION Here we report that TLS transcriptionally regulates the expression of oxidative stress protection genes. This regulation requires interaction with the transcriptional coactivator peroxisome proliferator activated receptor γ-coactivator 1α (PGC-1α), a master regulator of mitochondrial function that coordinately induces the expression of genes involved in detoxification of mitochondrial reactive oxygen species (ROS). Microarray gene expression analysis showed that TLS transcriptional activity is impaired in the absence of PGC-1α, and is thus largely dependent on PGC-1α. CONCLUSION These results suggest the existence of a regulatory circuit linking the control of ROS detoxification to the coordinated cross-talk between oxidative metabolism and the cellular response to genomic DNA damage.
Collapse
|
48
|
Hong T, Ning J, Yang X, Liu HY, Han J, Liu Z, Cao W. Fine-tuned regulation of the PGC-1α gene transcription by different intracellular signaling pathways. Am J Physiol Endocrinol Metab 2011; 300:E500-7. [PMID: 21156859 PMCID: PMC3064010 DOI: 10.1152/ajpendo.00225.2010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
It has previously been known that transcription of the PGC-1α gene can be either inhibited or stimulated by p38 MAP kinase (p38 MAPK). To determine whether p38 MAPK plays an inhibitory or stimulatory role in PGC-1α gene transcription, we further investigated the role of p38 MAPK in this study. Our results showed that the basal level of p38 MAPK phosphorylation was increased in gastrocnemius of mice under HFD and that p38 MAPK stimulated PGC-1α gene transcription in C(2)C(12) myotubes. Our results also provided new mechanisms in myotubes that the p38 MAPK-induced PGC-1α gene transcription was mediated by CREB. In exploring the role of the Akt-dependent insulin signaling on PGC-1α gene transcription, we found that the basal Akt-dependent signaling was increased in gastrocnemius of mice under HFD. The p38 MAPK-induced PGC-1α gene transcription was prevented by insulin. Insulin suppression of PGC-1α gene transcription was neutralized by overexpression of the constitutively nuclear form of FoxO1. Finally, we located three insulin response elements (IREs) in the PGC-1α promoter, and mutations of these IREs abolish or blunt activity of the PGC-1α promoter. Together, our results show that transcription of the PGC-1α gene is balanced by different intracellular signaling pathways.
Collapse
Affiliation(s)
- Tao Hong
- Translational Biology, The Hamner Institutes for Health Sciences, Research Triangle Park, NC 27709, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Afrose S, Hossain MS, Maki T, Tsujii H. Effects of karaya saponin and Rhodobacter capsulatus on yolk cholesterol in laying hens. Br Poult Sci 2010; 51:409-18. [PMID: 20680876 DOI: 10.1080/00071668.2010.495469] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
1. It has been reported that karaya saponin and Rhodobacter capsulatus individually have hypocholesterolaemic activity in laying hens. This study focuses on the effect of adding karaya saponin with R. capsulatus to hen's diet with regard to serum and egg yolk cholesterol and triglycerides. 2. A total of 56 Boris Brown laying hens were divided into 7 groups at 20 weeks of age. Combinations of 25, 50, 75 mg kg(-1) karaya saponin and R. capsulatus 200 and 400 mg kg(-1) were used as treatment groups. 3. After 8 weeks of supplementation, the effects of all the combinations of karaya saponin and R. capsulatus on serum and egg yolk cholesterol, triglycerides, and high-density lipoprotein (HDL)-cholesterol were greater than either karaya saponin or R. capsulatus alone. The combination of karaya saponin 50 mg kg(-1)+ R. capsulatus 400 mg kg(-1) exhibited the greatest reduction of serum (325%) and yolk (225%) cholesterol and the greatest increase of faecal, liver bile acids and yolk fatty acid (oleic, linoleic and linolenic) concentrations. In addition, egg production and yolk colour were significantly improved by the combined use of karaya saponin and R. capsulatus supplementation. 4. Therefore, the dietary supplementation of karaya saponin and R. capsulatus may lead to the production of a low-cholesterol egg, with production performance maintained at a standard level.
Collapse
Affiliation(s)
- S Afrose
- Shinshu University, Minamiminowa-mura, Japan
| | | | | | | |
Collapse
|
50
|
|