1
|
De Matteis C, Novielli F, Di Buduo E, Arconzo M, Gadaleta RM, Cariello M, Moschetta A, Crudele L. Atherogenic index of plasma identifies subjects with severe liver steatosis. Sci Rep 2025; 15:9136. [PMID: 40097487 PMCID: PMC11914574 DOI: 10.1038/s41598-025-93141-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025] Open
Abstract
The Atherogenic Index of Plasma (AIP), calculated by log (Triglycerides/HDL-C), has been proposed as a marker of atherogenic and cardiovascular risk. Atherosclerosis and Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) share some pathogenic features and may be considered clinical manifestations of Metabolic Syndrome. In this study, we aimed to investigate the role of increased AIP as a putative metabolic biomarker for MASLD. 1,496 individuals (49% males and 51% females) underwent clinical examination for Metabolic Syndrome at Internal Medicine Division "C. Frugoni" of University Hospital of Bari, Italy in the period between January 2016 and April 2024. Clinical history was recorded, and physical examination, anthropometric measures, biochemical assessment, and abdomen ultrasound were performed. In the overall population, AIP significantly correlated with fasting glycemia (FPG, r = 0.26, p < 0.0001), HbA1c (r = 0.20, p < 0.0001), LDL (r = 0.11, p < 0.0001) and total cholesterol (r = 0.09; p < 0.0001), and anthropometric measures of obesity BMI (r = 0.37, p < 0.0001) and Waist Circumference (r = 0.44; p < 0.001). We then investigated AIP values in patients with and without dysmetabolic conditions, finding that AIP significantly increased as steatosis worsened (p < 0.001). ROC curves identified an optimal cut-off of 0.31 for accurately diagnosing severe steatosis and AIP values above this cut-off discriminated patients with significantly increased (p < 0.0001) fasting glycemia, LDL, and waist circumference, and were strongly associated (p < 0.0001) with MASLD (LLR 85.3), type 2 diabetes (LLR 85.5), abdominal obesity (LLR 72.9), overweight (LLR 151.8), and systemic obesity (LLR 178.4). The risk for being diagnosed with such conditions was found to be even higher in the subpopulation of patients with severe liver steatosis. To validate our findings, we considered another cohort of patients with and without biopsy-proven liver steatosis (public dataset GSE89632), confirming that a significant increase (p < 0.001) in AIP values could be found in patients with liver steatosis compared to healthy controls. AIP can be considered a specific biomarker of fatty liver disease with high sensitivity for the diagnosis of the severe form of liver steatosis. Considering AIP in the evaluation of patients with liver steatosis may augment the accuracy for diagnosing metabolic impairment and MASLD.
Collapse
Affiliation(s)
- Carlo De Matteis
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare N. 11, 70124, Bari, Italy
| | - Fabio Novielli
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare N. 11, 70124, Bari, Italy
| | - Ersilia Di Buduo
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare N. 11, 70124, Bari, Italy
| | - Maria Arconzo
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare N. 11, 70124, Bari, Italy
| | - Raffaella Maria Gadaleta
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare N. 11, 70124, Bari, Italy
- INBB National Institute for Biostructure and Biosystems, Via Dei Carpegna, 19 - 00165, Roma, Italia
| | - Marica Cariello
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare N. 11, 70124, Bari, Italy
- INBB National Institute for Biostructure and Biosystems, Via Dei Carpegna, 19 - 00165, Roma, Italia
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare N. 11, 70124, Bari, Italy.
- INBB National Institute for Biostructure and Biosystems, Via Dei Carpegna, 19 - 00165, Roma, Italia.
| | - Lucilla Crudele
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare N. 11, 70124, Bari, Italy.
- INBB National Institute for Biostructure and Biosystems, Via Dei Carpegna, 19 - 00165, Roma, Italia.
| |
Collapse
|
2
|
Al Amaz S, Shahid MAH, Jha R, Mishra B. Prehatch thermal manipulation of embryos and posthatch baicalein supplementation increased liver metabolism, and muscle proliferation in broiler chickens. Poult Sci 2024; 103:104155. [PMID: 39216265 PMCID: PMC11402044 DOI: 10.1016/j.psj.2024.104155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
The exposure of broiler chickens to high ambient temperatures causes heat stress (HS), negatively affecting their health and production performance. To mitigate heat stress in broilers, various strategies, including dietary, managerial, and genetic interventions, have been extensively tested with varying degrees of efficacy. For sustainable broiler production, it is imperative to develop an innovative approach that effectively mitigates the adverse effects of HS. Our previous studies have provided valuable insights into the effects of prehatch embryonic thermal manipulation (TM) and posthatch baicalein supplementation on embryonic thermotolerance, metabolism, and posthatch growth performance. This follow-up study investigated the effect of these interventions on gluconeogenesis and lipid metabolism in the liver, as well as muscle proliferation and regeneration capacity in heat-stressed broiler chickens. A total of six-hundred fertile Cobb 500 eggs were incubated for 21 d. After candling, 238 eggs were subjected to TM at 38.5°C with 55% relative humidity (RH) from embryonic day (ED) 12 to 18. These eggs were transferred to the hatcher and kept at a standard temperature (37.5°C) from ED 19 to 21, while 236 eggs were incubated at a controlled temperature (37.5°C) till hatch. After hatching, 180 day-old chicks from both groups were raised in 36 pens treatment (n = 10 birds/pen, 6 replicates per treatment). The treatments were: 1) Control, 2) TM, 3) Control heat stress (CHS), 4) Thermal manipulation heat stress (TMHS), 5) Control heat stress supplement (CHSS), and 6) Thermal manipulation heat stress supplement (TMHSS). Baicalein was added to the treatment group diets starting from d 1. All birds were raised under the standard environment for 21 d, followed by chronic heat stress from d 22 to 35 (32-33 ⁰C for 8 h) in the CHS, TMHS, CHSS, and TMHSS groups. A thermoneutral (22-24⁰C) environment was maintained in the Control and TM groups. RH was constant (50 ± 5%) throughout the trial. In the liver, TM significantly increased (P < 0.05) IGF2 expression. Baicalein supplementation significantly increased (P < 0.05) HSF3, HSP70, SOD1, SOD2, TXN, PRARα, and GHR expression. Moreover, the combination of TM and baicalein supplementation significantly increased (P < 0.05) the expression of HSPH1, HSPB1, HSP90, LPL, and GHR. In the muscle, TM significantly increased (P < 0.05) HSF3 and Myf5 gene expression. TM and baicalein supplementation significantly increased (P < 0.05) the expression of MyoG and significantly (P < 0.05) decreased mTOR and PAX7. In conclusion, the prehatch TM of embryos and posthatch baicalein supplementation mitigated the deleterious effects of HS on broiler chickens by upregulating genes related to liver gluconeogenesis, lipid metabolism, and muscle proliferation.
Collapse
Affiliation(s)
- Sadid Al Amaz
- Department of Human Nutrition, Food and Animal Sciences, College of Tropical Agriculture and Human Resources, University of Hawai'i at Manoa, Honolulu, HI 96822
| | - Md Ahosanul Haque Shahid
- Department of Human Nutrition, Food and Animal Sciences, College of Tropical Agriculture and Human Resources, University of Hawai'i at Manoa, Honolulu, HI 96822
| | - Rajesh Jha
- Department of Human Nutrition, Food and Animal Sciences, College of Tropical Agriculture and Human Resources, University of Hawai'i at Manoa, Honolulu, HI 96822
| | - Birendra Mishra
- Department of Human Nutrition, Food and Animal Sciences, College of Tropical Agriculture and Human Resources, University of Hawai'i at Manoa, Honolulu, HI 96822.
| |
Collapse
|
3
|
Klemm RW, Carvalho P. Lipid Droplets Big and Small: Basic Mechanisms That Make Them All. Annu Rev Cell Dev Biol 2024; 40:143-168. [PMID: 39356808 DOI: 10.1146/annurev-cellbio-012624-031419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Lipid droplets (LDs) are dynamic storage organelles with central roles in lipid and energy metabolism. They consist of a core of neutral lipids, such as triacylglycerol, which is surrounded by a monolayer of phospholipids and specialized surface proteins. The surface composition determines many of the LD properties, such as size, subcellular distribution, and interaction with partner organelles. Considering the diverse energetic and metabolic demands of various cell types, it is not surprising that LDs are highly heterogeneous within and between cell types. Despite their diversity, all LDs share a common biogenesis mechanism. However, adipocytes have evolved specific adaptations of these basic mechanisms, enabling the regulation of lipid and energy metabolism at both the cellular and organismal levels. Here, we discuss recent advances in the understanding of both the general mechanisms of LD biogenesis and the adipocyte-specific adaptations controlling these fascinating organelles.
Collapse
Affiliation(s)
- Robin W Klemm
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom;
| | - Pedro Carvalho
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom;
| |
Collapse
|
4
|
Lei X, Chen H, Xu Y, Yang Z, Zhang L, Wang C, Du H. Serum isthmin-1 is a potential biomarker for metabolic dysfunction associated fatty liver disease in patients with metabolic syndrome and type 2 diabetes mellitus. BMJ Open Diabetes Res Care 2024; 12:e004514. [PMID: 39322582 PMCID: PMC11425935 DOI: 10.1136/bmjdrc-2024-004514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/12/2024] [Indexed: 09/27/2024] Open
Abstract
INTRODUCTION Metabolic dysfunction associated fatty liver disease (MAFLD) is a prevalent condition in patients with type 2 diabetes mellitus (T2DM). Isthmin-1 (ISM1) is an adipokine that promotes glucose uptake and improves glucose tolerance and hepatic steatosis. Although ISM1 has been shown to be associated with T2DM, its role in patients with MAFLD and metabolic syndrome (MetS) remains insufficiently examined. This study aimed to investigate the relationship between serum ISM1 and MAFLD in patients with T2DM and the potential involvement of MetS in this association. RESEARCH DESIGN AND METHODS A total of 250 participants were divided into four groups: 60 patients with T2DM and MAFLD, 60 with newly diagnosed T2DM, 60 with MAFLD, and 70 healthy controls. Serum ISM1 levels were measured using ELISA. The distribution of ISM1 concentration in the combined data was divided into quartiles, and the Cochran-Armitage trend test was performed to estimate the significant trends across increasing quartiles. RESULTS Compared with the controls, patients with coexisting MAFLD, MetS, and T2DM exhibited significantly elevated serum ISM1 concentrations. Serum ISM1 levels in the overweight/obese group were also higher than those in the lean group. Serum ISM1 levels were positively correlated with body mass index (BMI), uric acid, alanine aminotransferase, aspartate aminotransferase, total cholesterol (TC), low-density lipoprotein cholesterol, fasting insulin, and homeostasis model assessment of insulin resistance and negatively associated with age and high-density lipoprotein cholesterol (HDL-C). BMI, TC, and HDL-C were independently associated with serum ISM1 concentration. The relative risks for MAFLD, T2DM, and T2DM with MAFLD increased significantly with higher ISM1 quartiles. Furthermore, a positive correlation was observed between serum ISM1 levels and the number of MetS components, with the elevated plasma levels of ISM1 escalating the risk of developing MetS to some extent. CONCLUSIONS The combination of ISM1 with TG and UA was identified as the best predictive factor for diagnosing MAFLD and MetS, potentially due to their contribution to aggravating the metabolic state.
Collapse
Affiliation(s)
- Xiaohui Lei
- Department of Endocrinology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, Chongqing, China
| | - HaiYan Chen
- Department of Endocrinology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, Chongqing, China
| | - YuXin Xu
- Department of Endocrinology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, Chongqing, China
| | - Zhuoran Yang
- Department of Endocrinology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, Chongqing, China
| | - Lili Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, Chongqing, China
| | - Cong Wang
- Department of Endocrinology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, Chongqing, China
| | - Hu Du
- Department of Critical Care Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, Chongqing, China
| |
Collapse
|
5
|
Almohawes ZN, El-Kott A, Morsy K, Shati AA, El-Kenawy AE, Khalifa HS, Elsaid FG, Abd-Lateif AEKM, Abu-Zaiton A, Ebealy ER, Abdel-Daim MM, Ghanem RA, Abd-Ella EM. Salidroside inhibits insulin resistance and hepatic steatosis by downregulating miR-21 and subsequent activation of AMPK and upregulation of PPARα in the liver and muscles of high fat diet-fed rats. Arch Physiol Biochem 2024; 130:257-274. [PMID: 35061559 DOI: 10.1080/13813455.2021.2024578] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/15/2021] [Accepted: 12/27/2021] [Indexed: 02/06/2023]
Abstract
This study evaluated if salidroside (SAL) alleviates high-fat diet (HFD)-induced non-alcoholic fatty liver disease (NAFLD) by downregulating miR-21. Rats (n = 8/group) were treated for 12 weeks as normal diet (control/ND), ND + agmoir negative control (NC) (150 µg/kg), ND + SAL (300 mg/kg), HFD, HFD + SAL, HFD + compound C (an AMPK inhibitor) (200 ng/kg), HFD + SAL + NXT629 (a PPAR-α antagonist) (30 mg/kg), and HFD + SAL + miR-21 agomir (150 µg/kg). SAL improved glucose and insulin tolerance and preserved livers in HFD-fed rats. In ND and HFD-fed rats, SAL reduced levels of serum and hepatic lipids and the hepatic expression of SREBP1, SREBP2, fatty acid (FA) synthase, and HMGCOAR. It also activated hepatic Nrf2 and increased hepatic/muscular activity of AMPK and levels of PPARα. All effects afforded by SAL were prevented by CC, NXT629, and miR-21 agmoir. In conclusion, activation of AMPK and upregulation of PPARα mediate the anti-steatotic effect of SAL.
Collapse
Affiliation(s)
- Zakiah N Almohawes
- Biology Department, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Attalla El-Kott
- Biology Department, College of Science, King Khalid University, Abha, Saudi Arabia
- Zoology Department, College of Science, Damanhour University, Damanhour, Egypt
| | - Kareem Morsy
- Biology Department, College of Science, King Khalid University, Abha, Saudi Arabia
- Zoology Department, College of Science, Cairo University, Cairo, Egypt
| | - Ali A Shati
- Biology Department, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Ayman E El-Kenawy
- Pathology Department, College of Medicine, Taif University, Taif, Saudi Arabia
| | - Heba S Khalifa
- Zoology Department, College of Science, Damanhour University, Damanhour, Egypt
| | - Fahmy G Elsaid
- Biology Department, College of Science, King Khalid University, Abha, Saudi Arabia
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | | | | | - Eman R Ebealy
- Biology Department, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Mohamed M Abdel-Daim
- Pharmaceutical Sciences Department, Pharmacy Program, Batterjee Medical College, Jeddah, Saudi Arabia
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Reham A Ghanem
- Oral Biology Department, Faculty of Oral and Dental Medicine, Delta University for Science and Technology, Gamasa, Egypt
| | - Eman M Abd-Ella
- Zoology Department, College of Science, Fayoum University, Fayoum, Egypt
- Biology Department, College of Science and Art, Al-Baha University, Al-Mandaq, Saudi Arabia
| |
Collapse
|
6
|
Bo T, Gao L, Yao Z, Shao S, Wang X, Proud CG, Zhao J. Hepatic selective insulin resistance at the intersection of insulin signaling and metabolic dysfunction-associated steatotic liver disease. Cell Metab 2024; 36:947-968. [PMID: 38718757 DOI: 10.1016/j.cmet.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/22/2024] [Accepted: 04/09/2024] [Indexed: 06/26/2024]
Abstract
Insulin resistance (IR) is a major pathogenic factor in the progression of MASLD. In the liver, insulin suppresses gluconeogenesis and enhances de novo lipogenesis (DNL). During IR, there is a defect in insulin-mediated suppression of gluconeogenesis, but an unrestrained increase in hepatic lipogenesis persists. The mechanism of increased hepatic steatosis in IR is unclear and remains controversial. The key discrepancy is whether insulin retains its ability to directly regulate hepatic lipogenesis. Blocking insulin/IRS/AKT signaling reduces liver lipid deposition in IR, suggesting insulin can still regulate lipid metabolism; hepatic glucose metabolism that bypasses insulin's action may contribute to lipogenesis; and due to peripheral IR, other tissues are likely to impact liver lipid deposition. We here review the current understanding of insulin's action in governing different aspects of hepatic lipid metabolism under normal and IR states, with the purpose of highlighting the essential issues that remain unsettled.
Collapse
Affiliation(s)
- Tao Bo
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ling Gao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, China
| | - Zhenyu Yao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, China
| | - Shanshan Shao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, China
| | - Xuemin Wang
- Lifelong Health, South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA, Australia
| | - Christopher G Proud
- Lifelong Health, South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA, Australia.
| | - Jiajun Zhao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, China.
| |
Collapse
|
7
|
Kuriyama T, Ishibashi C, Kozawa J, Baden MY, Horii T, Niki A, Ozawa H, Hosokawa Y, Fujita Y, Sadahiro K, Satoh T, Hamaguchi T, Shimomura I. Effects of liraglutide on intrapancreatic fat deposition in patients with type 2 diabetes. Clin Nutr ESPEN 2024; 59:208-213. [PMID: 38220377 DOI: 10.1016/j.clnesp.2023.12.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/01/2023] [Accepted: 12/05/2023] [Indexed: 01/16/2024]
Abstract
BACKGROUND & AIMS Ectopic fat deposition is associated with worsening of glycemic control. This study was conducted to determine whether liraglutide reduces ectopic fat deposition, especially in pancreas, in patients with type 2 diabetes (T2D). METHODS We retrospectively recruited T2D patients who underwent abdominal unenhanced CT scans both before and after administration of liraglutide (N = 13) or glimepiride (N = 29). Using CT values of pancreas (P), liver (L) and spleen (S), we defined the indices of intrapancreatic and liver fat as P-S value and L-S value, respectively. Increase of each value suggests the reduction of each fat deposition. RESULTS The values of HbA1c (p = 0.0017) and body weight (p = 0.0081) decreased, and L-S (p = 0.0024) increased significantly after administration of liraglutide compared with those at baseline. Similarly, P-S tended to increase in the liraglutide group (p = 0.0547) and increased significantly in the liraglutide subgroup with fatty pancreas (p = 0.0303), defined as having baseline P-S less than -5. In the glimepiride group, P-S did not increase regardless of baseline P-S. Among patients with fatty pancreas, administration of liraglutide tended to be a significant factor for the change in P-S after adjustment for the change in HbA1c (p = 0.1090) and the change in visceral fat area (p = 0.1030). CONCLUSIONS Intrapancreatic fat deposition was decreased after treatment with liraglutide, but not glimepiride, in T2D patients with fatty pancreas. Liraglutide might reduce intrapancreatic fat deposition independently of decreases in HbA1c and visceral fat volume.
Collapse
Affiliation(s)
- Tsukasa Kuriyama
- Division of Diabetes, Department of Internal Medicine, Itami City Hospital, Itami, Hyogo, Japan; Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Chisaki Ishibashi
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Junji Kozawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan; Department of Diabetes Care Medicine, Graduate School of Medicine, Osaka University, Suita, Japan.
| | - Megu Y Baden
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan; Department of Lifestyle Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Tomomi Horii
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Akiko Niki
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Harutoshi Ozawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan; Department of Diabetes Care Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yoshiya Hosokawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yukari Fujita
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Katsuhiko Sadahiro
- Division of Diabetes, Department of Internal Medicine, Itami City Hospital, Itami, Hyogo, Japan
| | - Tomomi Satoh
- Division of Diabetes, Department of Internal Medicine, Itami City Hospital, Itami, Hyogo, Japan
| | - Tomoya Hamaguchi
- Division of Diabetes, Department of Internal Medicine, Itami City Hospital, Itami, Hyogo, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| |
Collapse
|
8
|
Uehara K, Lee WD, Stefkovich M, Biswas D, Santoleri D, Garcia Whitlock A, Quinn W, Coopersmith T, Creasy KT, Rader DJ, Sakamoto K, Rabinowitz JD, Titchenell PM. mTORC1 controls murine postprandial hepatic glycogen synthesis via Ppp1r3b. J Clin Invest 2024; 134:e173782. [PMID: 38290087 PMCID: PMC10977990 DOI: 10.1172/jci173782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/26/2024] [Indexed: 02/01/2024] Open
Abstract
In response to a meal, insulin drives hepatic glycogen synthesis to help regulate systemic glucose homeostasis. The mechanistic target of rapamycin complex 1 (mTORC1) is a well-established insulin target and contributes to the postprandial control of liver lipid metabolism, autophagy, and protein synthesis. However, its role in hepatic glucose metabolism is less understood. Here, we used metabolomics, isotope tracing, and mouse genetics to define a role for liver mTORC1 signaling in the control of postprandial glycolytic intermediates and glycogen deposition. We show that mTORC1 is required for glycogen synthase activity and glycogenesis. Mechanistically, hepatic mTORC1 activity promotes the feeding-dependent induction of Ppp1r3b, a gene encoding a phosphatase important for glycogen synthase activity whose polymorphisms are linked to human diabetes. Reexpression of Ppp1r3b in livers lacking mTORC1 signaling enhances glycogen synthase activity and restores postprandial glycogen content. mTORC1-dependent transcriptional control of Ppp1r3b is facilitated by FOXO1, a well characterized transcriptional regulator involved in the hepatic response to nutrient intake. Collectively, we identify a role for mTORC1 signaling in the transcriptional regulation of Ppp1r3b and the subsequent induction of postprandial hepatic glycogen synthesis.
Collapse
Affiliation(s)
- Kahealani Uehara
- Institute for Diabetes, Obesity, and Metabolism
- Biochemistry and Molecular Biophysics Graduate Group, and
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Won Dong Lee
- Lewis Sigler Institute for Integrative Genomics
- Department of Chemistry, and
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton, New Jersey, USA
| | | | - Dipsikha Biswas
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Dominic Santoleri
- Institute for Diabetes, Obesity, and Metabolism
- Biochemistry and Molecular Biophysics Graduate Group, and
| | | | | | | | - Kate Townsend Creasy
- Institute for Diabetes, Obesity, and Metabolism
- Department of Medicine, Division of Translational Medicine and Human Genetics, and
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel J. Rader
- Institute for Diabetes, Obesity, and Metabolism
- Department of Medicine, Division of Translational Medicine and Human Genetics, and
| | - Kei Sakamoto
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Joshua D. Rabinowitz
- Lewis Sigler Institute for Integrative Genomics
- Department of Chemistry, and
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton, New Jersey, USA
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Paul M. Titchenell
- Institute for Diabetes, Obesity, and Metabolism
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
9
|
Chen M, Lin Y, Dang Y, Xiao Y, Zhang F, Sun G, Jiang X, Zhang L, Du J, Duan S, Zhang X, Qin Z, Yang J, Liu K, Wu B. Reprogramming of rhythmic liver metabolism by intestinal clock. J Hepatol 2023; 79:741-757. [PMID: 37230230 DOI: 10.1016/j.jhep.2023.04.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 04/10/2023] [Accepted: 04/27/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND & AIMS Temporal oscillations in intestinal nutrient processing and absorption are coordinated by the local clock, which leads to the hypothesis that the intestinal clock has major impacts on shaping peripheral rhythms via diurnal nutritional signals. Here, we investigate the role of the intestinal clock in controlling liver rhythmicity and metabolism. METHODS Transcriptomic analysis, metabolomics, metabolic assays, histology, quantitative (q)PCR, and immunoblotting were performed with Bmal1-intestine-specific knockout (iKO), Rev-erba-iKO, and control mice. RESULTS Bmal1 iKO caused large-scale reprogramming of the rhythmic transcriptome of mouse liver with a limited effect on its clock. In the absence of intestinal Bmal1, the liver clock was resistant to entrainment by inverted feeding and a high-fat diet. Importantly, Bmal1 iKO remodelled diurnal hepatic metabolism by shifting to gluconeogenesis from lipogenesis during the dark phase, leading to elevated glucose production (hyperglycaemia) and insulin insensitivity. Conversely, Rev-erba iKO caused a diversion to lipogenesis from gluconeogenesis during the light phase, resulting in enhanced lipogenesis and an increased susceptibility to alcohol-related liver injury. These temporal diversions were attributed to disruption of hepatic SREBP-1c rhythmicity, which was maintained via gut-derived polyunsaturated fatty acids produced by intestinal FADS1/2 under the control of a local clock. CONCLUSIONS Our findings establish a pivotal role for the intestinal clock in dictating liver rhythmicity and diurnal metabolism, and suggest targeting intestinal rhythms as a new avenue for improving metabolic health. IMPACT AND IMPLICATIONS Our findings establish the centrality of the intestinal clock among peripheral tissue clocks, and associate liver-related pathologies with its malfunction. Clock modifiers in the intestine are shown to modulate liver metabolism with improved metabolic parameters. Such knowledge will help clinicians improve the diagnosis and treatment of metabolic diseases by incorporating intestinal circadian factors.
Collapse
Affiliation(s)
- Min Chen
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanke Lin
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongkang Dang
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yifei Xiao
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fugui Zhang
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guanghui Sun
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuejun Jiang
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Li Zhang
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianhao Du
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuyi Duan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaojian Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zifei Qin
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Jing Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Kaisheng Liu
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China.
| | - Baojian Wu
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
10
|
Muniz-Santos R, Lucieri-Costa G, de Almeida MAP, Moraes-de-Souza I, Brito MADSM, Silva AR, Gonçalves-de-Albuquerque CF. Lipid oxidation dysregulation: an emerging player in the pathophysiology of sepsis. Front Immunol 2023; 14:1224335. [PMID: 37600769 PMCID: PMC10435884 DOI: 10.3389/fimmu.2023.1224335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 06/30/2023] [Indexed: 08/22/2023] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by abnormal host response to infection. Millions of people are affected annually worldwide. Derangement of the inflammatory response is crucial in sepsis pathogenesis. However, metabolic, coagulation, and thermoregulatory alterations also occur in patients with sepsis. Fatty acid mobilization and oxidation changes may assume the role of a protagonist in sepsis pathogenesis. Lipid oxidation and free fatty acids (FFAs) are potentially valuable markers for sepsis diagnosis and prognosis. Herein, we discuss inflammatory and metabolic dysfunction during sepsis, focusing on fatty acid oxidation (FAO) alterations in the liver and muscle (skeletal and cardiac) and their implications in sepsis development.
Collapse
Affiliation(s)
- Renan Muniz-Santos
- Laboratory of Immunopharmacology, Department of Physiology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Giovanna Lucieri-Costa
- Laboratory of Immunopharmacology, Department of Physiology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Matheus Augusto P. de Almeida
- Neuroscience Graduate Program, Federal Fluminense University, Niteroi, Brazil
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Isabelle Moraes-de-Souza
- Laboratory of Immunopharmacology, Department of Physiology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Adriana Ribeiro Silva
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Cassiano Felippe Gonçalves-de-Albuquerque
- Laboratory of Immunopharmacology, Department of Physiology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
- Neuroscience Graduate Program, Federal Fluminense University, Niteroi, Brazil
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
11
|
Uehara K, Santoleri D, Whitlock AEG, Titchenell PM. Insulin Regulation of Hepatic Lipid Homeostasis. Compr Physiol 2023; 13:4785-4809. [PMID: 37358513 PMCID: PMC10760932 DOI: 10.1002/cphy.c220015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
The incidence of obesity, insulin resistance, and type II diabetes (T2DM) continues to rise worldwide. The liver is a central insulin-responsive metabolic organ that governs whole-body metabolic homeostasis. Therefore, defining the mechanisms underlying insulin action in the liver is essential to our understanding of the pathogenesis of insulin resistance. During periods of fasting, the liver catabolizes fatty acids and stored glycogen to meet the metabolic demands of the body. In postprandial conditions, insulin signals to the liver to store excess nutrients into triglycerides, cholesterol, and glycogen. In insulin-resistant states, such as T2DM, hepatic insulin signaling continues to promote lipid synthesis but fails to suppress glucose production, leading to hypertriglyceridemia and hyperglycemia. Insulin resistance is associated with the development of metabolic disorders such as cardiovascular and kidney disease, atherosclerosis, stroke, and cancer. Of note, nonalcoholic fatty liver disease (NAFLD), a spectrum of diseases encompassing fatty liver, inflammation, fibrosis, and cirrhosis, is linked to abnormalities in insulin-mediated lipid metabolism. Therefore, understanding the role of insulin signaling under normal and pathologic states may provide insights into preventative and therapeutic opportunities for the treatment of metabolic diseases. Here, we provide a review of the field of hepatic insulin signaling and lipid regulation, including providing historical context, detailed molecular mechanisms, and address gaps in our understanding of hepatic lipid regulation and the derangements under insulin-resistant conditions. © 2023 American Physiological Society. Compr Physiol 13:4785-4809, 2023.
Collapse
Affiliation(s)
- Kahealani Uehara
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dominic Santoleri
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anna E. Garcia Whitlock
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Paul M. Titchenell
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
12
|
Kamoshita K, Ishii KA, Tahira Y, Kikuchi A, Abuduwaili H, Tajima-Shirasaki N, Li Q, Takayama H, Matsumoto K, Takamura T. Insulin Suppresses Ubiquitination via the Deubiquitinating Enzyme Ubiquitin-Specific Protease 14, Independent of Proteasome Activity in H4IIEC3 Hepatocytes. J Pharmacol Exp Ther 2023; 385:5-16. [PMID: 36328485 DOI: 10.1124/jpet.122.001088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 08/16/2022] [Accepted: 09/09/2022] [Indexed: 11/06/2022] Open
Abstract
Ubiquitin-proteasome dysfunction contributes to obesity-related metabolic disorders, such as diabetes and fatty liver disease. However, the regulation of ubiquitin-proteasome activity by insulin remains to be elucidated. Here, we show that prolonged insulin stimulation activates proteasome function even though it reduces the ubiquitinated proteins in H4IIEC3 hepatocytes. Looking for a pathway by which insulin inhibits ubiquitination, we found that hepatic expression of ubiquitin-specific protease 14 (USP14) was upregulated in the liver of patients with insulin resistance. Indeed, the USP14-specific inhibitor IU1 canceled the insulin-mediated reduction of ubiquitinated proteins. Furthermore, insulin-induced endoplasmic reticulum (ER) stress, which was canceled by IU1, suggesting that USP14 activity is involved in insulin-induced ER stress. Co-stimulation with insulin and IU1 for 2 hours upregulated the nuclear translocation of the lipogenic transcription factor, sterol regulatory element binding protein-1c (SREBP-1c), upregulated the expression of the lipogenic gene, fatty acid synthase (Fasn), and repressed the gluconeogenic genes. In conclusion, insulin activates proteasome function even though it inhibits protein ubiquitination by activating USP14 in hepatocytes. USP14 activation by insulin inhibits mature SREBP-1c while upregulating ER stress and the expression of genes involved in gluconeogenesis. Further understanding mechanisms underlying the USP14 activation and its pleiotropic effects may lead to therapeutic development for obesity-associated metabolic disorders, such as diabetes and fatty liver disease. SIGNIFICANCE STATEMENT: This study shows that insulin stimulation inhibits ubiquitination by activating USP14, independent of its effect on proteasome activity in hepatocytes. USP14 also downregulates the nuclear translocation of the lipogenic transcription factor SREBP-1c and upregulates the expression of genes involved in gluconeogenesis. Since USP14 is upregulated in the liver of insulin-resistant patients, understanding mechanisms underlying the USP14 activation and its pleiotropic effects will help develop treatments for metabolic disorders such as diabetes and fatty liver.
Collapse
Affiliation(s)
- Kyoko Kamoshita
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Kiyo-Aki Ishii
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yumiko Tahira
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Akihiro Kikuchi
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Halimulati Abuduwaili
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Natsumi Tajima-Shirasaki
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Qifang Li
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Hiroaki Takayama
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Kunio Matsumoto
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Toshinari Takamura
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| |
Collapse
|
13
|
Icaritin, a metabolite of Icarrin, Alleviates non-alcoholic fatty liver disease via inhibition of lipogenesis and ER stress. Eur J Pharmacol 2022; 937:175378. [DOI: 10.1016/j.ejphar.2022.175378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/13/2022]
|
14
|
Park JE, Son J, Seo Y, Han JS. HM-Chromanone Ameliorates Hyperglycemia and Dyslipidemia in Type 2 Diabetic Mice. Nutrients 2022; 14:1951. [PMID: 35565920 PMCID: PMC9101766 DOI: 10.3390/nu14091951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/26/2022] [Accepted: 05/05/2022] [Indexed: 02/04/2023] Open
Abstract
The effects of (E)-5-hydroxy-7-methoxy-3-(2-hydroxybenzyl)-4-chromanone (HMC) on hyperglycemia and dyslipidemia were investigated in diabetic mice. Mice were separated into three groups: db/db, rosiglitazone and HMC. Blood glucose or glycosylated hemoglobin values in HMC-treated mice were significantly lower compared to db/db mice. Total cholesterol, LDL-cholesterol, and triglyceride values were lower, and HDL-C levels were higher, in the HMC group compared to the diabetic and rosiglitazone groups. HMC markedly increased IRS-1Tyr612, AktSer473 and PI3K levels and plasma membrane GLUT4 levels in skeletal muscle, suggesting improved insulin resistance. HMC also significantly stimulated AMPKThr172 and PPARα in the liver, and ameliorated dyslipidemia by inhibiting SREBP-1c and FAS. Consequently, HMC reduced hyperglycemia by improving the expression of insulin-resistance-related genes and improved dyslipidemia by regulating fatty acid synthase and oxidation-related genes in db/db mice. Therefore, HMC could ameliorate hyperglycemia and dyslipidemia in type 2 diabetic mice.
Collapse
Affiliation(s)
- Jae Eun Park
- Department of Food Science and Nutrition, Pusan National University, Busan 46241, Korea;
| | - Jaemin Son
- Division of Marine Bioscience, Ocean Science & Technology School, Korea Maritime and Ocean University, Busan 49112, Korea; (J.S.); (Y.S.)
| | - Youngwan Seo
- Division of Marine Bioscience, Ocean Science & Technology School, Korea Maritime and Ocean University, Busan 49112, Korea; (J.S.); (Y.S.)
| | - Ji Sook Han
- Department of Food Science and Nutrition, Pusan National University, Busan 46241, Korea;
| |
Collapse
|
15
|
Lee G, Kim YY, Jang H, Han JS, Nahmgoong H, Park YJ, Han SM, Cho C, Lim S, Noh JR, Oh WK, Lee CH, Kim S, Kim JB. SREBP1c-PARP1 axis tunes anti-senescence activity of adipocytes and ameliorates metabolic imbalance in obesity. Cell Metab 2022; 34:702-718.e5. [PMID: 35417665 DOI: 10.1016/j.cmet.2022.03.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 12/28/2021] [Accepted: 03/23/2022] [Indexed: 01/10/2023]
Abstract
Emerging evidence indicates that the accretion of senescent cells is linked to metabolic disorders. However, the underlying mechanisms and metabolic consequences of cellular senescence in obesity remain obscure. In this study, we found that obese adipocytes are senescence-susceptible cells accompanied with genome instability. Additionally, we discovered that SREBP1c may play a key role in genome stability and senescence in adipocytes by modulating DNA-damage responses. Unexpectedly, SREBP1c interacted with PARP1 and potentiated PARP1 activity during DNA repair, independent of its canonical lipogenic function. The genetic depletion of SREBP1c accelerated adipocyte senescence, leading to immune cell recruitment into obese adipose tissue. These deleterious effects provoked unhealthy adipose tissue remodeling and insulin resistance in obesity. In contrast, the elimination of senescent adipocytes alleviated adipose tissue inflammation and improved insulin resistance. These findings revealed distinctive roles of SREBP1c-PARP1 axis in the regulation of adipocyte senescence and will help decipher the metabolic significance of senescence in obesity.
Collapse
Affiliation(s)
- Gung Lee
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Ye Young Kim
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Hagoon Jang
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Ji Seul Han
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Hahn Nahmgoong
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Yoon Jeong Park
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Sang Mun Han
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Changyun Cho
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 08826, South Korea
| | - Sangsoo Lim
- Bioinformatics Institute, Seoul National University, Seoul 08826, South Korea
| | - Jung-Ran Noh
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, University of Science and Technology, Yuseong-gu, Daejeon 34141, South Korea
| | - Won Keun Oh
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, University of Science and Technology, Yuseong-gu, Daejeon 34141, South Korea
| | - Sun Kim
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 08826, South Korea; Bioinformatics Institute, Seoul National University, Seoul 08826, South Korea; Department of Computer Science and Engineering, Institute of Engineering Research, Seoul National University, Seoul 08826, South Korea
| | - Jae Bum Kim
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, South Korea.
| |
Collapse
|
16
|
Abstract
Hepatic lipogenesis is fine-tuned by mechanistic target of rapamycin (mTOR) signaling.
Collapse
Affiliation(s)
- Henry N Ginsberg
- Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Arya Mani
- Department of Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
17
|
Lu X, Ding Y, Liu H, Sun M, Chen C, Yang Y, Wang H. The Role of Hydrogen Sulfide Regulation of Autophagy in Liver Disorders. Int J Mol Sci 2022; 23:ijms23074035. [PMID: 35409395 PMCID: PMC8999478 DOI: 10.3390/ijms23074035] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 02/01/2023] Open
Abstract
Autophagy is a complex process of degradation of senescent or dysfunctional organelles in cells. Dysfunctional autophagy is associated with many diseases such as cancers, immune dysfunction, and aging. Hydrogen sulfide (H2S) is considered to be the third gas signal molecule after nitrous oxide and carbon monoxide. In recent years, H2S has been found to have a variety of important biological functions, and plays an important role in a variety of physiological and pathological processes. In this review, we review the recent role and mechanism of H2S in regulating autophagy in liver disorders, in order to provide a basis for further research in the future.
Collapse
Affiliation(s)
- Xueqin Lu
- Institute of Nursing and Health, School of Nursing and Health, Henan University, Jinming Avenue, Kaifeng 475004, China; (X.L.); (Y.D.); (C.C.)
| | - Yueming Ding
- Institute of Nursing and Health, School of Nursing and Health, Henan University, Jinming Avenue, Kaifeng 475004, China; (X.L.); (Y.D.); (C.C.)
| | - Huiyang Liu
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (H.L.); (M.S.); (Y.Y.)
| | - Mengyao Sun
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (H.L.); (M.S.); (Y.Y.)
| | - Chaoran Chen
- Institute of Nursing and Health, School of Nursing and Health, Henan University, Jinming Avenue, Kaifeng 475004, China; (X.L.); (Y.D.); (C.C.)
| | - Yihan Yang
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (H.L.); (M.S.); (Y.Y.)
| | - Honggang Wang
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (H.L.); (M.S.); (Y.Y.)
- Correspondence:
| |
Collapse
|
18
|
Dam TV, Toft NI, Grøntved L. Cell-Type Resolved Insights into the Cis-Regulatory Genome of NAFLD. Cells 2022; 11:870. [PMID: 35269495 PMCID: PMC8909044 DOI: 10.3390/cells11050870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/27/2022] [Accepted: 02/28/2022] [Indexed: 11/20/2022] Open
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) is increasing rapidly, and unmet treatment can result in the development of hepatitis, fibrosis, and liver failure. There are difficulties involved in diagnosing NAFLD early and for this reason there are challenges involved in its treatment. Furthermore, no drugs are currently approved to alleviate complications, a fact which highlights the need for further insight into disease mechanisms. NAFLD pathogenesis is associated with complex cellular changes, including hepatocyte steatosis, immune cell infiltration, endothelial dysfunction, hepatic stellate cell activation, and epithelial ductular reaction. Many of these cellular changes are controlled by dramatic changes in gene expression orchestrated by the cis-regulatory genome and associated transcription factors. Thus, to understand disease mechanisms, we need extensive insights into the gene regulatory mechanisms associated with tissue remodeling. Mapping cis-regulatory regions genome-wide is a step towards this objective and several current and emerging technologies allow detection of accessible chromatin and specific histone modifications in enriched cell populations of the liver, as well as in single cells. Here, we discuss recent insights into the cis-regulatory genome in NAFLD both at the organ-level and in specific cell populations of the liver. Moreover, we highlight emerging technologies that enable single-cell resolved analysis of the cis-regulatory genome of the liver.
Collapse
Affiliation(s)
| | | | - Lars Grøntved
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense, Denmark; (T.V.D.); (N.I.T.)
| |
Collapse
|
19
|
Uehara K, Sostre-Colón J, Gavin M, Santoleri D, Leonard KA, Jacobs RL, Titchenell PM. Activation of Liver mTORC1 Protects Against NASH via Dual Regulation of VLDL-TAG Secretion and De Novo Lipogenesis. Cell Mol Gastroenterol Hepatol 2022; 13:1625-1647. [PMID: 35240344 PMCID: PMC9046248 DOI: 10.1016/j.jcmgh.2022.02.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND & AIMS Dysregulation of liver lipid metabolism is associated with the development and progression of nonalcoholic fatty liver disease, a spectrum of liver diseases including nonalcoholic steatohepatitis (NASH). In the liver, insulin controls lipid homeostasis by increasing triglyceride (TAG) synthesis, suppressing fatty acid oxidation, and enhancing TAG export via very low-density lipoproteins. Downstream of insulin signaling, the mechanistic target of rapamycin complex 1 (mTORC1), is a key regulator of lipid metabolism. Here, we define the role of hepatic mTORC1 activity in mouse models of NASH and investigate the mTORC1-dependent mechanisms responsible for protection against liver damage in NASH. METHODS Utilizing 2 rodent NASH-promoting diets, we demonstrate that hepatic mTORC1 activity was reduced in mice with NASH, whereas under conditions of insulin resistance and benign fatty liver, mTORC1 activity was elevated. To test the beneficial effects of hepatic mTORC1 activation in mouse models of NASH, we employed an acute, liver-specific knockout model of TSC1 (L-TSC-KO), a negative regulator of mTORC1. RESULTS L-TSC-KO mice are protected from and have improved markers of NASH including reduced steatosis, decreased circulating transaminases, and reduced expression of inflammation and fibrosis genes. Mechanistically, protection from hepatic inflammation and fibrosis by constitutive mTORC1 activity occurred via promotion of the phosphatidylcholine synthesizing enzyme, CCTα, and enhanced very low-density lipoprotein-triglyceride export. Additionally, activation of mTORC1 protected from hepatic steatosis via negative feedback of the mTORC2-AKT-FOXO-SREBP1c lipogenesis axis. CONCLUSIONS Collectively, this study identifies a protective role for liver mTORC1 signaling in the initiation and progression of NASH in mice via dual control of lipid export and synthesis.
Collapse
Affiliation(s)
- Kahealani Uehara
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jaimarie Sostre-Colón
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Matthew Gavin
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dominic Santoleri
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kelly-Ann Leonard
- Department of Agricultural, Food and Nutritional Science Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - René L Jacobs
- Department of Agricultural, Food and Nutritional Science Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Paul M Titchenell
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
20
|
Szymczak-Pajor I, Miazek K, Selmi A, Balcerczyk A, Śliwińska A. The Action of Vitamin D in Adipose Tissue: Is There the Link between Vitamin D Deficiency and Adipose Tissue-Related Metabolic Disorders? Int J Mol Sci 2022; 23:956. [PMID: 35055140 PMCID: PMC8779075 DOI: 10.3390/ijms23020956] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/09/2022] [Accepted: 01/10/2022] [Indexed: 12/11/2022] Open
Abstract
Adipose tissue plays an important role in systemic metabolism via the secretion of adipocytokines and storing and releasing energy. In obesity, adipose tissue becomes dysfunctional and characterized by hypertrophied adipocytes, increased inflammation, hypoxia, and decreased angiogenesis. Although adipose tissue is one of the major stores of vitamin D, its deficiency is detective in obese subjects. In the presented review, we show how vitamin D regulates numerous processes in adipose tissue and how their dysregulation leads to metabolic disorders. The molecular response to vitamin D in adipose tissue affects not only energy metabolism and adipokine and anti-inflammatory cytokine production via the regulation of gene expression but also genes participating in antioxidant defense, adipocytes differentiation, and apoptosis. Thus, its deficiency disturbs adipocytokines secretion, metabolism, lipid storage, adipogenesis, thermogenesis, the regulation of inflammation, and oxidative stress balance. Restoring the proper functionality of adipose tissue in overweight or obese subjects is of particular importance in order to reduce the risk of developing obesity-related complications, such as cardiovascular diseases and diabetes. Taking into account the results of experimental studies, it seemed that vitamin D may be a remedy for adipose tissue dysfunction, but the results of the clinical trials are not consistent, as some of them show improvement and others no effect of this vitamin on metabolic and insulin resistance parameters. Therefore, further studies are required to evaluate the beneficial effects of vitamin D, especially in overweight and obese subjects, due to the presence of a volumetric dilution of this vitamin among them.
Collapse
Affiliation(s)
- Izabela Szymczak-Pajor
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland;
| | - Krystian Miazek
- Institute of Applied Radiation Chemistry, Faculty of Chemistry, Lodz University of Technology, 15 Wroblewskiego, 93-590 Lodz, Poland;
| | - Anna Selmi
- Department of Molecular Biophysics, University of Lodz, 141/143 Pomorska, 90-236 Lodz, Poland; (A.S.); (A.B.)
| | - Aneta Balcerczyk
- Department of Molecular Biophysics, University of Lodz, 141/143 Pomorska, 90-236 Lodz, Poland; (A.S.); (A.B.)
| | - Agnieszka Śliwińska
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland;
| |
Collapse
|
21
|
Insulin-Responsive Transcription Factors. Biomolecules 2021; 11:biom11121886. [PMID: 34944530 PMCID: PMC8699568 DOI: 10.3390/biom11121886] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/04/2021] [Accepted: 12/09/2021] [Indexed: 12/12/2022] Open
Abstract
The hormone insulin executes its function via binding and activating of the insulin receptor, a receptor tyrosine kinase that is mainly expressed in skeletal muscle, adipocytes, liver, pancreatic β-cells, and in some areas of the central nervous system. Stimulation of the insulin receptor activates intracellular signaling cascades involving the enzymes extracellular signal-regulated protein kinase-1/2 (ERK1/2), phosphatidylinositol 3-kinase, protein kinase B/Akt, and phospholipase Cγ as signal transducers. Insulin receptor stimulation is correlated with multiple physiological and biochemical functions, including glucose transport, glucose homeostasis, food intake, proliferation, glycolysis, and lipogenesis. This review article focuses on the activation of gene transcription as a result of insulin receptor stimulation. Signal transducers such as protein kinases or the GLUT4-induced influx of glucose connect insulin receptor stimulation with transcription. We discuss insulin-responsive transcription factors that respond to insulin receptor activation and generate a transcriptional network executing the metabolic functions of insulin. Importantly, insulin receptor stimulation induces transcription of genes encoding essential enzymes of glycolysis and lipogenesis and inhibits genes encoding essential enzymes of gluconeogenesis. Overall, the activation or inhibition of insulin-responsive transcription factors is an essential aspect of orchestrating a wide range of insulin-induced changes in the biochemistry and physiology of insulin-responsive tissues.
Collapse
|
22
|
Beta vulgaris L. (Beetroot) Methanolic Extract Prevents Hepatic Steatosis and Liver Damage in T2DM Rats by Hypoglycemic, Insulin-Sensitizing, Antioxidant Effects, and Upregulation of PPARα. BIOLOGY 2021; 10:biology10121306. [PMID: 34943221 PMCID: PMC8698622 DOI: 10.3390/biology10121306] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/04/2021] [Accepted: 12/06/2021] [Indexed: 12/15/2022]
Abstract
Simple Summary Beetroot is one of the most consumable plants across the world. Previous studies have shown many health benefits of beetroot, with evidence of having potent hypoglycemic, antioxidant, and anti-inflammatory effects. The data obtained from this study further confirmed this effect in streptozotocin-diabetic animals. They showed the ability of methanolic beetroot extract to prevent the associated hepatic oxidative stress, inflammation, steatosis, and dyslipidaemia. However, the protection mechanisms involve, at least, upregulation of endogenous antioxidants, anti-apoptotic Bcl2, and PPARα. Abstract The present study examined if methanolic beetroot extract (BE) could prevent dyslipidemia and hepatic steatosis and damage in a type-2 diabetes mellitus (T2DM) rat model and studied some mechanisms of action. T2DM was induced in adult male Wistar rats by a low single dose of streptozotocin (STZ) (35 mg/kg, i.p) and a high-fat diet (HFD) feeding for 5 weeks. Control or T2DM rats then continued on standard or HFDs for another 12 weeks and were treated with the vehicle or BE (250 or 500 mg/kg). BE, at both doses, significantly improved liver structure and reduced hepatic lipid accumulation in the livers of T2DM rats. They also reduced body weight gain, serum glucose, insulin levels, serum and hepatic levels of cholesterol, triglycerides, free fatty acids, and serum levels of low-density lipoproteins in T2DM rats. In concomitant, they significantly reduced serum levels of aspartate and alanine aminotransferases, hepatic levels of malondialdehyde, tumor-necrosis factor-α, interleukin-6, and mRNA of Bax, cleaved caspase-3, and SREBP1/2. However, both doses of BE significantly increased hepatic levels of total glutathione, superoxide dismutase, and mRNA levels of Bcl2 and PPARα in the livers of both the control and T2DM rats. All of these effects were dose-dependent and more profound with doses of 500 mg/kg. In conclusion, chronic feeding of BE to STZ/HFD-induced T2DM in rats prevents hepatic steatosis and liver damage by its hypoglycemic and insulin-sensitizing effects and its ability to upregulate antioxidants and PPARα.
Collapse
|
23
|
SREBP-1c and lipogenesis in the liver: an update1. Biochem J 2021; 478:3723-3739. [PMID: 34673919 DOI: 10.1042/bcj20210071] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 12/13/2022]
Abstract
Sterol Regulatory Element Binding Protein-1c is a transcription factor that controls the synthesis of lipids from glucose in the liver, a process which is of utmost importance for the storage of energy. Discovered in the early nineties by B. Spiegelman and by M. Brown and J. Goldstein, it has generated more than 5000 studies in order to elucidate its mechanism of activation and its role in physiology and pathology. Synthetized as a precursor found in the membranes of the endoplasmic reticulum, it has to be exported to the Golgi and cleaved by a mechanism called regulated intramembrane proteolysis. We reviewed in 2002 its main characteristics, its activation process and its role in the regulation of hepatic glycolytic and lipogenic genes. We particularly emphasized that Sterol Regulatory Element Binding Protein-1c is the mediator of insulin effects on these genes. In the present review, we would like to update these informations and focus on the response to insulin and to another actor in Sterol Regulatory Element Binding Protein-1c activation, the endoplasmic reticulum stress.
Collapse
|
24
|
The Interplay between Insulin Resistance, Inflammation, Oxidative Stress, Base Excision Repair and Metabolic Syndrome in Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2021; 22:ijms222011128. [PMID: 34681787 PMCID: PMC8537238 DOI: 10.3390/ijms222011128] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 02/06/2023] Open
Abstract
One of the most common chronic liver disorders, affecting mainly people in Western countries, is nonalcoholic fatty liver disease (NAFLD). Unfortunately, its pathophysiological mechanism is not fully understood, and no dedicated treatment is available. Simple steatosis can lead to nonalcoholic steatohepatitis and even to fibrosis, cancer, and cirrhosis of the liver. NAFLD very often occurs in parallel with type 2 diabetes mellitus and in obese people. Furthermore, it is much more likely to develop in patients with metabolic syndrome (MS), whose criteria include abdominal obesity, elevated blood triacylglycerol level, reduced high-density lipoprotein cholesterol level, increased blood pressure, and high fasting glucose. An important phenomenon in MS is also insulin resistance (IR), which is very common in NAFLD. Liver IR and NAFLD development are linked through an interaction between the accumulation of free fatty acids, hepatic inflammation, and increased oxidative stress. The liver is particularly exposed to elevated levels of reactive oxygen species due to a large number of mitochondria in hepatocytes. In these organelles, the main DNA repair pathway is base excision repair (BER). The present article will illustrate how impairment of BER may be related to the development of NAFLD.
Collapse
|
25
|
Shatoor AS, Al Humayed S, Almohiy HM. Astaxanthin attenuates hepatic steatosis in high-fat diet-fed rats by suppressing microRNA-21 via transactivation of nuclear factor erythroid 2-related factor 2. J Physiol Biochem 2021; 78:151-168. [PMID: 34651285 DOI: 10.1007/s13105-021-00850-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 09/29/2021] [Indexed: 02/08/2023]
Abstract
This study examined whether astaxanthin (ASX) could alleviate hepatic steatosis in rats fed a high-fat diet (HFD) by modulating the nuclear factor erythroid 2-related factor 2 (Nrf2)/miR-21 axis. Rats (n = 8/group) were fed either a standard diet (3.8 kcal/g; 10% fat) or HFD (4.6 kcal/g; 40% fat) and treated orally with either the vehicle or ASX (6 mg/kg) daily for 8 days. Another group was fed HFD and treated with ASX and brusatol (an Nrf2 inhibitor) (2 mg/kg/twice per week/i.p.). ASX prevented the gain in body and liver weights and attenuated hepatic lipid accumulation in HFD-fed rats. In the control and HFD-fed rats, ASX did not affect food intake, serum free fatty acid (FFA) content, and glucose and insulin levels and tolerance. However, serum triglyceride (TG), cholesterol, and low-density lipoprotein-cholesterol levels; hepatic levels of TGs and FFAs; and hepatic levels of Srebp1, Srebp2, HMGCR, and fatty acid synthase mRNAs and miR-21 were reduced and the mRNA levels of Pparα were significantly increased in both the groups. These effects were associated with a reduction in the hepatic levels of reactive oxygen species, malondialdehyde, tumor necrosis factor-α, and interlukin-6 as well as an increase in superoxide dismutase levels, total glutathione content, and nuclear levels and activity of Nrf2. miR-21 levels were strongly correlated with the nuclear activity of Nrf2. Brusatol completely reversed the effects of ASX. In conclusion, ASX prevents hepatic steatosis mainly by transactivating Nrf2 and is associated with the suppression of miR-21 and Srebp1/2 and upregulation of Pparα expression.
Collapse
Affiliation(s)
- Abdullah S Shatoor
- Department of Medicine, Cardiology Section, College of Medicine, King Khalid University (KKU), Abha, Saudi Arabia.
| | - Suliman Al Humayed
- Department of Internal Medicine, College of Medicine, King Khalid University (KKU), Abha, Saudi Arabia
| | - Hussain M Almohiy
- Depatrtment of Radiology Science, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
26
|
Abstract
The molecular mechanisms of cellular insulin action have been the focus of much investigation since the discovery of the hormone 100 years ago. Insulin action is impaired in metabolic syndrome, a condition known as insulin resistance. The actions of the hormone are initiated by binding to its receptor on the surface of target cells. The receptor is an α2β2 heterodimer that binds to insulin with high affinity, resulting in the activation of its tyrosine kinase activity. Once activated, the receptor can phosphorylate a number of intracellular substrates that initiate discrete signaling pathways. The tyrosine phosphorylation of some substrates activates phosphatidylinositol-3-kinase (PI3K), which produces polyphosphoinositides that interact with protein kinases, leading to activation of the kinase Akt. Phosphorylation of Shc leads to activation of the Ras/MAP kinase pathway. Phosphorylation of SH2B2 and of Cbl initiates activation of G proteins such as TC10. Activation of Akt and other protein kinases produces phosphorylation of a variety of substrates, including transcription factors, GTPase-activating proteins, and other kinases that control key metabolic events. Among the cellular processes controlled by insulin are vesicle trafficking, activities of metabolic enzymes, transcriptional factors, and degradation of insulin itself. Together these complex processes are coordinated to ensure glucose homeostasis.
Collapse
|
27
|
Jiang Z, Zhao M, Voilquin L, Jung Y, Aikio MA, Sahai T, Dou FY, Roche AM, Carcamo-Orive I, Knowles JW, Wabitsch M, Appel EA, Maikawa CL, Camporez JP, Shulman GI, Tsai L, Rosen ED, Gardner CD, Spiegelman BM, Svensson KJ. Isthmin-1 is an adipokine that promotes glucose uptake and improves glucose tolerance and hepatic steatosis. Cell Metab 2021; 33:1836-1852.e11. [PMID: 34348115 PMCID: PMC8429235 DOI: 10.1016/j.cmet.2021.07.010] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 06/02/2021] [Accepted: 07/09/2021] [Indexed: 02/07/2023]
Abstract
With the increasing prevalence of type 2 diabetes and fatty liver disease, there is still an unmet need to better treat hyperglycemia and hyperlipidemia. Here, we identify isthmin-1 (Ism1) as an adipokine and one that has a dual role in increasing adipose glucose uptake while suppressing hepatic lipid synthesis. Ism1 ablation results in impaired glucose tolerance, reduced adipose glucose uptake, and reduced insulin sensitivity, demonstrating an endogenous function for Ism1 in glucose regulation. Mechanistically, Ism1 activates a PI3K-AKT signaling pathway independently of the insulin and insulin-like growth factor receptors. Notably, while the glucoregulatory function is shared with insulin, Ism1 counteracts lipid accumulation in the liver by switching hepatocytes from a lipogenic to a protein synthesis state. Furthermore, therapeutic dosing of recombinant Ism1 improves diabetes in diet-induced obese mice and ameliorates hepatic steatosis in a diet-induced fatty liver mouse model. These findings uncover an unexpected, bioactive protein hormone that might have simultaneous therapeutic potential for diabetes and fatty liver disease.
Collapse
Affiliation(s)
- Zewen Jiang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Meng Zhao
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Laetitia Voilquin
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yunshin Jung
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mari A Aikio
- Department of Cell Biology, Harvard Medical School and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Tanushi Sahai
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Florence Y Dou
- Department of Cell Biology, Harvard Medical School and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Alexander M Roche
- Department of Cell Biology, Harvard Medical School and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Ivan Carcamo-Orive
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joshua W Knowles
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Prevention Research Center, Stanford University, Stanford, CA 94305, USA
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, University Medical Center Ulm, Ulm, Germany
| | - Eric A Appel
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA; Department of Pediatrics (Endocrinology), Stanford University, Stanford, CA 94305, USA
| | - Caitlin L Maikawa
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Joao Paulo Camporez
- Department of Physiology, Ribeirao Preto School of Medicine, University of Sao Paulo, Sao Paulo, Brazil; Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Gerald I Shulman
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA; Department of Cellular and Molecular Physiology and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Linus Tsai
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Evan D Rosen
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Bruce M Spiegelman
- Department of Cell Biology, Harvard Medical School and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Katrin J Svensson
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
28
|
A potential probiotic bacterium for antipsychotic-induced metabolic syndrome: mechanisms underpinning how Akkermansia muciniphila subtype improves olanzapine-induced glucose homeostasis in mice. Psychopharmacology (Berl) 2021; 238:2543-2553. [PMID: 34046717 DOI: 10.1007/s00213-021-05878-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 05/17/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Olanzapine (OLZ) is one of the most effective atypical antipsychotics but is associated with severe metabolic side effects, in which the gut microbiota plays an important role. Akkermansia muciniphila (A. muciniphila; Akk), a Gram-negative anaerobic bacterium in the intestine, can potentially improve metabolic syndrome. OBJECTIVE This study investigated the effect and underlying mechanisms of an A. muciniphila subtype (A. muciniphilasub; Akksub) on OLZ-induced metabolic dysfunction in lean and obese mice. METHODS C57BL/6 female mice were fed a high-fat diet to induce obesity or normal chow for 8 weeks before OLZ treatment for 16 weeks. During the treatment period, mice in each group were orally administrated A. muciniphilasub. Weight gain, glucose and lipid metabolism, and inflammation were evaluated. RESULTS A. muciniphilasub decreased OLZ-related weight gain only at week 16 in lean mice and significantly alleviated OLZ-induced hyperglycemia irrespective of diet. This was accompanied by reduced levels of glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxykinase (PEPCK)-key enzymes in hepatic gluconeogenesis-and OLZ-associated insulin resistance. Moreover, OLZ-induced increases in serum interleukin (IL)-6 and tumor necrosis factor (TNF)-α levels were improved by A. muciniphilasub in both obese and lean mice. OLZ did not increase serum lipid levels or hepatic fat accumulation. CONCLUSIONS A. muciniphilasub improves OLZ-related hyperglycemia via regulation of G6Pase and PEPCK levels and insulin resistance. Moreover, A. muciniphilasub alleviates systemic inflammation caused by OLZ. A. muciniphilasub is a promising probiotic treatment for OLZ-induced metabolic dysfunction.
Collapse
|
29
|
Park G, Jung S, Wellen KE, Jang C. The interaction between the gut microbiota and dietary carbohydrates in nonalcoholic fatty liver disease. Exp Mol Med 2021; 53:809-822. [PMID: 34017059 PMCID: PMC8178320 DOI: 10.1038/s12276-021-00614-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/24/2021] [Indexed: 02/04/2023] Open
Abstract
Imbalance between fat production and consumption causes various metabolic disorders. Nonalcoholic fatty liver disease (NAFLD), one such pathology, is characterized by abnormally increased fat synthesis and subsequent fat accumulation in hepatocytes1,2. While often comorbid with obesity and insulin resistance, this disease can also be found in lean individuals, suggesting specific metabolic dysfunction2. NAFLD has become one of the most prevalent liver diseases in adults worldwide, but its incidence in both children and adolescents has also markedly increased in developed nations3,4. Progression of this disease into nonalcoholic steatohepatitis (NASH), cirrhosis, liver failure, and hepatocellular carcinoma in combination with its widespread incidence thus makes NAFLD and its related pathologies a significant public health concern. Here, we review our understanding of the roles of dietary carbohydrates (glucose, fructose, and fibers) and the gut microbiota, which provides essential carbon sources for hepatic fat synthesis during the development of NAFLD.
Collapse
Affiliation(s)
- Grace Park
- Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | - Sunhee Jung
- Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
30
|
Abstract
Mammals undergo regular cycles of fasting and feeding that engage dynamic transcriptional responses in metabolic tissues. Here we review advances in our understanding of the gene regulatory networks that contribute to hepatic responses to fasting and feeding. The advent of sequencing and -omics techniques have begun to facilitate a holistic understanding of the transcriptional landscape and its plasticity. We highlight transcription factors, their cofactors, and the pathways that they impact. We also discuss physiological factors that impinge on these responses, including circadian rhythms and sex differences. Finally, we review how dietary modifications modulate hepatic gene expression programs.
Collapse
Affiliation(s)
- Lara Bideyan
- Department of Pathology and Laboratory Medicine, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA.,Department of Biological Chemistry, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Rohith Nagari
- Department of Pathology and Laboratory Medicine, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA.,Department of Biological Chemistry, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA.,Department of Biological Chemistry, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
31
|
Moldavski O, Zushin PJH, Berdan CA, Van Eijkeren RJ, Jiang X, Qian M, Ory DS, Covey DF, Nomura DK, Stahl A, Weiss EJ, Zoncu R. 4β-Hydroxycholesterol is a prolipogenic factor that promotes SREBP1c expression and activity through the liver X receptor. J Lipid Res 2021; 62:100051. [PMID: 33631213 PMCID: PMC8042401 DOI: 10.1016/j.jlr.2021.100051] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/06/2021] [Accepted: 02/12/2021] [Indexed: 12/16/2022] Open
Abstract
Oxysterols are oxidized derivatives of cholesterol that play regulatory roles in lipid biosynthesis and homeostasis. How oxysterol signaling coordinates different lipid classes such as sterols and triglycerides remains incompletely understood. Here, we show that 4β-hydroxycholesterol (HC) (4β-HC), a liver and serum abundant oxysterol of poorly defined functions, is a potent and selective inducer of the master lipogenic transcription factor, SREBP1c, but not the related steroidogenic transcription factor SREBP2. By correlating tracing of lipid synthesis with lipogenic gene expression profiling, we found that 4β-HC acts as a putative agonist for the liver X receptor (LXR), a sterol sensor and transcriptional regulator previously linked to SREBP1c activation. Unique among the oxysterol agonists of the LXR, 4β-HC induced expression of the lipogenic program downstream of SREBP1c and triggered de novo lipogenesis both in primary hepatocytes and in the mouse liver. In addition, 4β-HC acted in parallel to insulin-PI3K-dependent signaling to stimulate triglyceride synthesis and lipid-droplet accumulation. Thus, 4β-HC is an endogenous regulator of de novo lipogenesis through the LXR-SREBP1c axis.
Collapse
Affiliation(s)
- Ofer Moldavski
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA; The Paul F. Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA, USA; Cardiovascular Research Institute, UCSF, San Francisco, CA, USA
| | - Peter-James H Zushin
- Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA, USA
| | - Charles A Berdan
- Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA, USA
| | - Robert J Van Eijkeren
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA; The Paul F. Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA, USA
| | - Xuntian Jiang
- Diabetic Cardiovascular Disease Center, Washington University School of Medicine, St Louis, MO, USA
| | - Mingxing Qian
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Daniel S Ory
- Diabetic Cardiovascular Disease Center, Washington University School of Medicine, St Louis, MO, USA
| | - Douglas F Covey
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Daniel K Nomura
- Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA, USA
| | - Andreas Stahl
- Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA, USA
| | - Ethan J Weiss
- Cardiovascular Research Institute, UCSF, San Francisco, CA, USA
| | - Roberto Zoncu
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA; The Paul F. Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
32
|
Li D, Ikaga R, Ogawa H, Yamazaki T. Different expressions of clock genes in fatty liver induced by high-sucrose and high-fat diets. Chronobiol Int 2021; 38:762-778. [PMID: 33612041 DOI: 10.1080/07420528.2021.1889579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Sucrose consumption can cause obesity and nonalcoholic fatty liver disease (NAFLD). NAFLD is associated with the disruption of circadian rhythms. We compared the alterations in NAFLD circadian rhythms induced by a high-sucrose diet (HSD) with those induced by a high-fat diet (HFD) in mice. After 8 weeks of feeding, the liver triglyceride level was increased by HSD feeding and by HFD feeding. In the liver of HSD-fed mice, the amplitude of Rorγ and the mesor (time series 24 h mean value based on the distribution of values across the cycle of the circadian rhythm) of Rorγ and Per2 were increased in comparison to those of control-diet fed mice. Compared with the HFD-fed mice, the HSD-fed mice showed increased circadian amplitude of variation in Rorγ, Per2, Cry1, and Cry2 and mesors of Rorγ, Per2, and Cry1 in the liver. Rorγ appeared to play critical roles in the entrainment of HSD into the liver circadian system, and the increased expressions of Crys and Per2 might disrupt circadian rhythms. Thus, disruption of circadian rhythms by HSD and HFD may accelerate the accumulation of liver lipid through different mechanisms.
Collapse
Affiliation(s)
- Dongyang Li
- Department of Nutritional Science, National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo, Japan.,The Graduate School of Humanities and Sciences, Ochanomizu University, Tokyo, Japan
| | - Reina Ikaga
- Department of Nutritional Science, National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo, Japan
| | - Haruko Ogawa
- The Graduate School of Humanities and Sciences, and Institute for Human Life Innovation, Ochanomizu University, Tokyo, Japan
| | - Tomomi Yamazaki
- Department of Nutritional Science, National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo, Japan
| |
Collapse
|
33
|
Shochat C, Wang Z, Mo C, Nelson S, Donaka R, Huang J, Karasik D, Brotto M. Deletion of SREBF1, a Functional Bone-Muscle Pleiotropic Gene, Alters Bone Density and Lipid Signaling in Zebrafish. Endocrinology 2021; 162:5929645. [PMID: 33068391 PMCID: PMC7745669 DOI: 10.1210/endocr/bqaa189] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Indexed: 12/30/2022]
Abstract
Through a genome-wide analysis of bone mineral density (BMD) and muscle mass, identification of a signaling pattern on 17p11.2 recognized the presence of sterol regulatory element-binding factor 1 (SREBF1), a gene responsible for the regulation of lipid homeostasis. In conjunction with lipid-based metabolic functions, SREBF1 also codes for the protein, SREBP-1, a transcription factor known for its role in adipocyte differentiation. We conducted a quantitative correlational study. We established a zebrafish (ZF) SREBF1 knockout (KO) model and used a targeted customized lipidomics approach to analyze the extent of SREBF1 capabilities. For lipidomics profiling, we isolated the dorsal muscles of wild type (WT) and KO fishes, and we performed liquid chromatography-tandem mass spectrometry screening assays of these samples. In our analysis, we profiled 48 lipid mediators (LMs) derived from various essential polyunsaturated fatty acids to determine potential targets regulated by SREBF1, and we found that the levels of 11,12 epoxyeicosatrienoic acid (11,12-EET) were negatively associated with the number of SREBF1 alleles (P = 0.006 for a linear model). We also compared gene expression between KO and WT ZF by genome-wide RNA-sequencing. Significantly enriched pathways included fatty acid elongation, linoleic acid metabolism, arachidonic acid metabolism, adipocytokine signaling, and DNA replication. We discovered trends indicating that BMD in adult fish was significantly lower in the KO than in the WT population (P < 0.03). These studies reinforce the importance of lipidomics investigation by detailing how the KO of SREBF1 affects both BMD and lipid-signaling mediators, thus confirming the importance of SREBF1 for musculoskeletal homeostasis.
Collapse
Affiliation(s)
- Chen Shochat
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Zhiying Wang
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas at Arlington-UTA, Arlington, Texas
| | - Chenglin Mo
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas at Arlington-UTA, Arlington, Texas
| | - Sarah Nelson
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas at Arlington-UTA, Arlington, Texas
| | | | - Jian Huang
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas at Arlington-UTA, Arlington, Texas
| | - David Karasik
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
- Correspondence: David Karasik, Azrieli Faculty of Medicine, Bar-Ilan university, Safed, 1311502, Israel. E-mail:
| | - Marco Brotto
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas at Arlington-UTA, Arlington, Texas
| |
Collapse
|
34
|
Effects of dietary carbohydrate sources on lipid metabolism and SUMOylation modification in the liver tissues of yellow catfish. Br J Nutr 2020; 124:1241-1250. [PMID: 32600495 DOI: 10.1017/s0007114520002408] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Dysregulation in hepatic lipid synthesis by excess dietary carbohydrate intake is often relevant with the occurrence of fatty liver; therefore, the thorough understanding of the regulation of lipid deposition and metabolism seems crucial to search for potential regulatory targets. In the present study, we examined TAG accumulation, lipid metabolism-related gene expression, the enzyme activities of lipogenesis-related enzymes, the protein levels of transcription factors or genes involving lipogenesis in the livers of yellow catfish fed five dietary carbohydrate sources, such as glucose, maize starch, sucrose, potato starch and dextrin, respectively. Generally speaking, compared with other carbohydrate sources, dietary glucose promoted TAG accumulation, up-regulated lipogenic enzyme activities and gene expressions, and down-regulated mRNA expression of genes involved in lipolysis and small ubiquitin-related modifier (SUMO) modification pathways. Further studies found that sterol regulatory element binding protein 1 (SREBP1), a key transcriptional factor relevant to lipogenic regulation, was modified by SUMO1. Mutational analyses found two important sites for SUMOylation modification (K254R and K264R) in SREBP1. Mutant SREBP lacking lysine 264 up-regulated the transactivation capacity on an SREBP-responsive promoter. Glucose reduced the SUMOylation level of SREBP1 and promoted the protein expression of SREBP1 and its target gene stearoyl-CoA desaturase 1 (SCD1), indicating that SUMOylation of SREBP1 mediated glucose-induced hepatic lipid metabolism. Our study elucidated the molecular mechanism of dietary glucose increasing hepatic lipid deposition and found that the SREBP-dependent transactivation was regulated by SUMO1 modification, which served as a new target for the transcriptional programmes governing lipid metabolism.
Collapse
|
35
|
Sun LF, Yang YL, Xiao TX, Li MX, Zhang JV. Removal of DHT can relieve polycystic ovarian but not metabolic abnormalities in DHT-induced hyperandrogenism in mice. Reprod Fertil Dev 2020; 31:1597-1606. [PMID: 31142430 DOI: 10.1071/rd18459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 04/24/2019] [Indexed: 12/30/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine disorder with a high prevalence in women of childbearing age. To date, there is no method of efficiently diagnosing PCOS and curing it completely because its pathomechanism remains unclear. Here, we investigated whether metabolic abnormalities maintain the hyperandrogenism and PCOS-like ovaries and whether the symptoms induced by excess androgen are treatable. We ceased the abnormal dihydrotestosterone (DHT) stimulation to determine changes in PCOS-like mice. After ceasing DHT stimulation, the ovarian morphology and gene expression recovered from the DHT-stimulated status. However, after cessation of DHT stimulation, the hypertrophy of adipose tissues and hepatic steatosis were not significantly restored, and fat accumulation-related gene expression and serum metabolic markers in the mice were altered. These findings showed that the reproductive dysfunction was obviously relieved, but because the metabolic abnormalities were not relieved after the cessation of excess androgen for 30 days, it appears that the latter may not maintain the former.
Collapse
Affiliation(s)
- Li-Feng Sun
- Research Laboratory for Reproductive Health, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ya-Li Yang
- Research Laboratory for Reproductive Health, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Tian-Xia Xiao
- Research Laboratory for Reproductive Health, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Meng-Xia Li
- Research Laboratory for Reproductive Health, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jian V Zhang
- Research Laboratory for Reproductive Health, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; and Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; and Corresponding author.
| |
Collapse
|
36
|
Li YZ, Di Cristofano A, Woo M. Metabolic Role of PTEN in Insulin Signaling and Resistance. Cold Spring Harb Perspect Med 2020; 10:a036137. [PMID: 31964643 PMCID: PMC7397839 DOI: 10.1101/cshperspect.a036137] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Phosphatase and tensin homolog (PTEN) is most prominently known for its function in tumorigenesis. However, a metabolic role of PTEN is emerging as a result of its altered expression in type 2 diabetes (T2D), which results in impaired insulin signaling and promotion of insulin resistance during the pathogenesis of T2D. PTEN functions in regulating insulin signaling across different organs have been identified. Through the use of a variety of models, such as tissue-specific knockout (KO) mice and in vitro cell cultures, PTEN's role in regulating insulin action has been elucidated across many cell types. Herein, we will review the recent advancements in the understanding of PTEN's metabolic functions in each of the tissues and cell types that contribute to regulating systemic insulin sensitivity and discuss how PTEN may represent a promising therapeutic strategy for treatment or prevention of T2D.
Collapse
Affiliation(s)
- Yu Zhe Li
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5G 2M9, Canada
| | - Antonio Di Cristofano
- Department of Developmental and Molecular Biology and Medicine (Oncology), Albert Einstein College of Medicine and Albert Einstein Cancer Center, Bronx, New York 10461, USA
| | - Minna Woo
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5G 2M9, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario M5G 2M9, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, University Health Network/Mount Sinai Hospital, University of Toronto, Toronto, Ontario M5G 2C4, Canada
| |
Collapse
|
37
|
Mukai Y, Kataoka S, Sato S. Sorghum (Sorghum bicolor) Extract Affects Plasma Lipid Metabolism and Hepatic Macrophage Infiltration in Diabetic Rats. CURRENT NUTRITION & FOOD SCIENCE 2020. [DOI: 10.2174/1573401315666190114153933] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Chronic hyperglycemia is known to be a high-risk factor for progressive
chronic liver diseases, such as abnormal lipid metabolism. The activation of AMP-activated protein
kinase (AMPK) has a beneficial effect on dyslipidemia. Polyphenols derived from various plants are
involved in AMPK activation.
Objective:
We investigated the effects of polyphenol-containing sorghum (Sorghum bicolor) extract
(SE) on plasma lipid metabolism and macrophage infiltration, and measured the expression and
phosphorylation of AMPK and acetyl-CoA carboxylase (ACC) in diabetic rat livers.
Methods:
Streptozotocin-induced diabetic rats received 0, 50, or 250 mg/kg of SE orally for 4 weeks.
Blood chemistry, total and phosphorylated protein levels of AMPK and ACC, sterol regulatory element-
binding protein-1c (SREBP-1c) mRNA and protein levels, and macrophage infiltration in the
livers were examined.
Results:
Plasma glucose and triacylglycerol levels, which were increased in the untreated diabetic
rats, were significantly lower in the 250 mg/kg SE-treated diabetic rats. AMPK and ACC phosphorylation
levels were significantly increased in the 250 mg/kg SE-treated diabetic rats compared with
those in the untreated rats. There was no difference in the hepatic expression of SREBP-1c between
the diabetic rat groups. Macrophage infiltration in the liver was suppressed by 250 mg/kg of SEtreatment.
Conclusion:
These data suggest that SE treatment may affect plasma lipid metabolism and chronic
inflammation by upregulating phosphorylation of AMPK and ACC in diabetic rat livers.
Collapse
Affiliation(s)
- Yuuka Mukai
- Department of Food Hygiene and Function, School of Nutrition and Dietetics, Faculty of Health and Social Work, Kanagawa University of Human Services, Kanagawa, Japan
| | - Saori Kataoka
- Department of Nutrition, Aomori University of Health and Welfare, Aomori, Japan
| | - Shin Sato
- Department of Nutrition, Aomori University of Health and Welfare, Aomori, Japan
| |
Collapse
|
38
|
Fatty liver diseases, mechanisms, and potential therapeutic plant medicines. Chin J Nat Med 2020; 18:161-168. [PMID: 32245585 DOI: 10.1016/s1875-5364(20)30017-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Indexed: 02/07/2023]
Abstract
The liver is an important metabolic organ and controls lipid, glucose and energy metabolism. Dysruption of hepatic lipid metabolism is often associated with fatty liver diseases, including nonalcoholic fatty liver disease (NAFLD), alcoholic fatty liver diseases (AFLD) and hyperlipidemia. Recent studies have uncovered the contribution of hormones, transcription factors, and inflammatory cytokines to the pathogenesis of dyslipidemia and fatty liver diseases. Moreover, a significant amount of effort has been put to examine the mechanisms underlying the potential therapeutic effects of many natural plant products on fatty liver diseases and metabolic diseases. We review the current understanding of insulin, thyroid hormone and inflammatory cytokines in regulating hepatic lipid metabolism, focusing on several essential transcription regulators, such as Sirtuins (SIRTs), Forkhead box O (FoxO), Sterol-regulatory element-binding proteins (SREBPs). We also discuss a few representative natural products with promising thereapeutic effects on fatty liver disease and dyslipidemia.
Collapse
|
39
|
Yahaghi L, Yaghmaei P, Hayati-Roodbari N, Irani S, Ebrahim-Habibi A. Betanin effect on PPAR-α and SREBP-1c expression in NMRI mice model of steatohepatitis with fibrosis. Physiol Int 2020; 107:67-81. [DOI: 10.1556/2060.2020.00001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 12/20/2019] [Indexed: 11/19/2022]
Abstract
AbstractPurposeBetanin is a betacyanin with antioxidant and anti-inflammatory activities whose effects were investigated in a nonalcoholic steatohepatitis (NASH) model.Main methodsNinety-six male naval medical research institute (NMRI) mice were divided into eight groups (n = 12) including normal control, high fat diet (HFD), Sham, and positive control treated with trans-chalcone. Three experimental groups were treated with 5 mg/kg, 10 mg/kg or 20 mg/kg betanin, and a betanin protective group was also defined.ResultsFour weeks of HFD treatment resulted in steatohepatitis with associated fibrosis. Significant increase was observed in serum levels of triglycerides (TG), total cholesterol (TC), glucose, insulin, leptin, liver enzymes, malondialdehyde (MDA), furthermore insulin resistance and (sterol regulatory element-binding protein-1c) SREBP-1c were detected. Levels of high-density lipoprotein cholesterol (HDL-C), adiponectin, superoxide dismutase (SOD), catalase (CAT), and PPAR-α (peroxisome proliferator-activated receptor-α) considerably decreased. Treatment by betanin, particularly the 20 mg/kg dosage, attenuated these changes.ConclusionBetanin is a potential treating agent of steatohepatitis and works through up-regulation of PPAR-α, down-regulation of SREBP-1c, modification of adipokine levels and modulation of lipid profile.
Collapse
Affiliation(s)
- L. Yahaghi
- 1Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Parichehreh Yaghmaei
- 1Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - N. Hayati-Roodbari
- 1Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - S. Irani
- 1Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - A. Ebrahim-Habibi
- 2Biosensor Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- 3Endocrinology and Metabolism Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
40
|
Vergani L. Fatty Acids and Effects on In Vitro and In Vivo Models of Liver Steatosis. Curr Med Chem 2019; 26:3439-3456. [PMID: 28521680 DOI: 10.2174/0929867324666170518101334] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/14/2017] [Accepted: 03/14/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Fatty liver, or steatosis, is a condition of excess accumulation of lipids, mainly under form of triglycerides (TG), in the liver, and it is the hallmark of non-alcoholic fatty liver disease (NAFLD). NAFLD is the most common liver disorder world-wide and it has frequently been associated with obesity, hyperlipidemia and insulin resistance. Free fatty acids (FA) are the major mediators of hepatic steatosis; patients with NAFLD have elevated levels of circulating FA that correlate with disease severity. METHODS Steatosis is a reversible condition that can be resolved with changed behaviors, or that can progress towards more severe liver damages such as steatohepatitis (NASH), fibrosis and cirrhosis. In NAFLD, FA of exogenous or endogenous origin accumulate in the hepatocytes and trigger liver damages. Excess TG are stored in cytosolic lipid droplets (LDs) that are dynamic organelles acting as hubs for lipid metabolism. RESULTS In the first part of this review, we briefly reassumed the main classes of FA and their chemical classification as a function of the presence and number of double bonds, their metabolic pathways and effects on human health. Then, we summarized the main genetic and diet-induced animal models of NAFLD, as well as the cellular models of NAFLD. CONCLUSIONS In recent years, both the diet-induced animal models of NAFLD as well as the cellular models of NAFLD have found ever more application to investigate the mechanisms involved in NAFLD, and we referred to their advantages and disadvantages.
Collapse
Affiliation(s)
- Laura Vergani
- DISTAV, Department of Earth, Environment and Life Sciences, University of Genova, Italy
| |
Collapse
|
41
|
Wang Y, Buyse J, Courousse N, Tesseraud S, Métayer-Coustard S, Berri C, Schallier S, Everaert N, Collin A. Effects of sex and fasting/refeeding on hepatic AMPK signaling in chickens (Gallus gallus). Comp Biochem Physiol A Mol Integr Physiol 2019; 240:110606. [PMID: 31676410 DOI: 10.1016/j.cbpa.2019.110606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/10/2019] [Accepted: 10/18/2019] [Indexed: 11/16/2022]
Abstract
The alpha-1 isoform of chicken AMPK situates on the Z-chromosome, in contrast, the other isoforms in birds and the mammalian AMPKα1 are located on the autosomes. The present study aimed to investigate the role of hepatic AMPK signaling in adaptation to nutritional status and the potential sex-specific response in chickens. Hepatic genes and proteins were compared between the two sexes immediately after hatching. From 20d of age, chicks from each sex received feed treatments: Control was fed ad libitum; Fasted was starved for 24 h; Refed was fed for 4 h after a 24 h fasting. As a result, hepatic AMPKα1 mRNA level in males was significantly higher at both ages compared to females, due to the presence of Z-chromosomes. However, this did not make this kinase "male-bias" as it was eventually compensated at a translational level, which was not reported in previous studies. The protein levels and activation of AMPKα were even lower in newly-hatched male compared to female chicks, accompanied with a higher FAS and SREBP-1 gene expressions. Accordingly, hepatic G6PC2 mRNA levels in males were significantly lower associated with lower plasma glucose levels after hatching. Fasting activated hepatic AMPK, which in turn inhibited gene expression of GS, FAS and SREBP-1, and stimulated the downstream G6PC2 in both sexes. These changes recovered after refeeding. In conclusion, AMPK plays a role in adaptation to nutritional environment for both sexes. The Z-linked AMPK did not exert a sex-specific signaling, due to a "translational compensation" of AMPKα1.
Collapse
Affiliation(s)
- Yufeng Wang
- Laboratory of Livestock Physiology, Department of Biosystems, KU Leuven, Kasteelpark Arenberg 30, 3001 Leuven, Belgium
| | - Johan Buyse
- Laboratory of Livestock Physiology, Department of Biosystems, KU Leuven, Kasteelpark Arenberg 30, 3001 Leuven, Belgium.
| | | | | | | | - Cécile Berri
- BOA, INRA, Université de Tours, 37380 Nouzilly, France
| | - Seline Schallier
- Laboratory of Livestock Physiology, Department of Biosystems, KU Leuven, Kasteelpark Arenberg 30, 3001 Leuven, Belgium
| | - Nadia Everaert
- Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, TERRA Teaching and Research Centre, University of Liège, Passage des Déportés 2, 5030 Gembloux, Belgium
| | - Anne Collin
- BOA, INRA, Université de Tours, 37380 Nouzilly, France
| |
Collapse
|
42
|
Presby DM, Checkley LA, Jackman MR, Higgins JA, Jones KL, Giles ED, Houck JA, Webb PG, Steig AJ, Johnson GC, Rudolph MC, MacLean PS. Regular exercise potentiates energetically expensive hepatic de novo lipogenesis during early weight regain. Am J Physiol Regul Integr Comp Physiol 2019; 317:R684-R695. [PMID: 31553623 DOI: 10.1152/ajpregu.00074.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Exercise is a potent facilitator of long-term weight loss maintenance (WLM), whereby it decreases appetite and increases energy expenditure beyond the cost of the exercise bout. We have previously shown that exercise may amplify energy expenditure through energetically expensive nutrient deposition. Therefore, we investigated the effect of exercise on hepatic de novo lipogenesis (DNL) during WLM and relapse to obesity. Obese rats were calorically restricted with (EX) or without (SED) treadmill exercise (1 h/day, 6 days/wk, 15 m/min) to induce and maintain weight loss. After 6 wk of WLM, subsets of WLM-SED and WLM-EX rats were allowed ad libitum access to food for 1 day to promote relapse (REL). An energy gap-matched group of sedentary, relapsing rats (REL-GM) were provided a diet matched to the positive energy imbalance of the REL-EX rats. During relapse, exercise increased enrichment of hepatic DN-derived lipids and induced hepatic molecular adaptations favoring DNL compared with the gap-matched controls. In the liver, compared with both REL-SED and REL-GM rats, REL-EX rats had lower hepatic expression of genes required for cholesterol biosynthesis; greater hepatic expression of genes that mediate very low-density lipoprotein synthesis and secretion; and greater mRNA expression of Cyp27a1, which encodes an enzyme involved in the biosynthesis of bile acids. Altogether, these data provide compelling evidence that the liver has an active role in exercise-mediated potentiation of energy expenditure during early relapse.
Collapse
Affiliation(s)
- David M Presby
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - L Allyson Checkley
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Matthew R Jackman
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Janine A Higgins
- Department of Pediatrics, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Kenneth L Jones
- Department of Pediatrics, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Erin D Giles
- Department of Nutrition and Food Science at Texas A&M University, College Station, Texas
| | - Julie A Houck
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Patricia G Webb
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Amy J Steig
- Center for Human Nutrition at the University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Ginger C Johnson
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Michael C Rudolph
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Paul S MacLean
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado.,Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado.,Center for Human Nutrition at the University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
43
|
Laskowski D, Andersson G, Humblot P, Sirard MA, Sjunnesson Y, Ferreira CR, Pirro V, Båge R. Lipid profile of bovine blastocysts exposed to insulin during in vitro oocyte maturation. Reprod Fertil Dev 2019; 30:1253-1266. [PMID: 29655403 DOI: 10.1071/rd17248] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 03/01/2018] [Indexed: 01/22/2023] Open
Abstract
Insulin is a key hormone with important functions in energy metabolism and is involved in the regulation of reproduction. Hyperinsulinaemia is known to impair fertility (for example, in obese mothers); therefore, we aimed to investigate the impact of elevated insulin concentrations during the sensitive period of oocyte maturation on gene expression and lipid profiles of the bovine Day-8 embryo. Two different insulin concentrations were used during in vitro oocyte maturation (INS10=10µgmL-1 and INS0.1=0.1µgmL-1) in order to observe possible dose-dependent effects or thresholds for hyperinsulinaemia in vitro. By investigating gene expression patterns by an mRNA microarray in combination with lipid profile analysis by desorption electrospray ionisation-mass spectrometry (DESI-MS) of embryos derived from insulin-treated oocytes, we gained further insights regarding molecular responses of embryos to insulin provocation during the first days of development. Lipid metabolism appeared to be influenced on multiple levels according to gene expression results but the profiles collected in positive-ion mode by DESI-MS (showing mostly ubiquinone, cholesteryl esters and triacylglycerols) did not differ significantly from controls. There are parallels in follicular development of ruminants and humans that make this bovine model relevant for comparative research on early human embryonic development during hyperinsulinaemia.
Collapse
Affiliation(s)
- Denise Laskowski
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, PO Box 7054, SE-750 07 Uppsala, Sweden
| | - Göran Andersson
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, PO Box 7023, SE-750 07 Uppsala, Sweden
| | - Patrice Humblot
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, PO Box 7054, SE-750 07 Uppsala, Sweden
| | - Marc-André Sirard
- Departement des Sciences Animales, Centre de Recherche en Biologie de la Reproduction, Pavillon Des Services, Local 2732, University Laval, Québec G1V 0A6, Canada
| | - Ylva Sjunnesson
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, PO Box 7054, SE-750 07 Uppsala, Sweden
| | - Christina R Ferreira
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907-2084, USA
| | - Valentina Pirro
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907-2084, USA
| | - Renée Båge
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, PO Box 7054, SE-750 07 Uppsala, Sweden
| |
Collapse
|
44
|
SREBP1-dependent de novo fatty acid synthesis gene expression is elevated in malignant melanoma and represents a cellular survival trait. Sci Rep 2019; 9:10369. [PMID: 31316083 PMCID: PMC6637239 DOI: 10.1038/s41598-019-46594-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 07/01/2019] [Indexed: 02/07/2023] Open
Abstract
de novo fatty acid biosynthesis (DNFA) is a hallmark adaptation of many cancers that supports survival, proliferation, and metastasis. Here we elucidate previously unexplored aspects of transcription regulation and clinical relevance of DNFA in cancers. We show that elevated expression of DNFA genes is characteristic of many tumor types and correlates with poor prognosis, especially in melanomas. Elevated DNFA gene expression depends on the SREBP1 transcription factor in multiple melanoma cell lines. SREBP1 predominantly binds to the transcription start sites of DNFA genes, regulating their expression by recruiting RNA polymerase II to promoters for productive transcription elongation. We find that SREBP1-regulated DNFA represents a survival trait in melanoma cells, regardless of proliferative state and oncogenic mutation status. Indeed, malignant melanoma cells exhibit elevated DNFA gene expression after the BRAF/MEK signaling pathway is blocked (e.g. by BRAF inhibitors), and DNFA expression remains higher in melanoma cells resistant to vemurafenib treatment than in untreated cells. Accordingly, DNFA pathway inhibition, whether by direct targeting of SREBP1 with antisense oligonucleotides, or through combinatorial effects of multiple DNFA enzyme inhibitors, exerts potent cytotoxic effects on both BRAFi-sensitive and -resistant melanoma cells. Altogether, these results implicate SREBP1 and DNFA enzymes as enticing therapeutic targets in melanomas.
Collapse
|
45
|
Ortega-Prieto P, Postic C. Carbohydrate Sensing Through the Transcription Factor ChREBP. Front Genet 2019; 10:472. [PMID: 31275349 PMCID: PMC6593282 DOI: 10.3389/fgene.2019.00472] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 05/01/2019] [Indexed: 12/23/2022] Open
Abstract
Carbohydrate response element binding protein (ChREBP) is a carbohydrate-signaling transcription factor that in the past years has emerged as a central metabolic regulator. ChREBP expression is mostly abundant in active sites of de novo lipogenesis including liver and white and brown adipose tissues. ChREBP is also expressed in pancreatic islets, small intestine and to a lesser extent in the kidney and the brain. In response to glucose, ChREBP undergoes several post-translational modifications (PTMs) (phosphorylation, acetylation and/or O-GlcNAcylation) that will either modulate its cellular location, stability and/or its transcriptional activity. ChREBPβ is a shorter isoform of ChREBP that was first described in adipose tissue and later found to be expressed in other sites including liver and pancreatic β cells. ChREBPβ lacks an important regulatory inhibitory domain, known as LID (low glucose inhibitory domain), in its N-terminal domain and is therefore reported as a highly active isoform. In this review, we recapitulate a recent progress concerning the mechanisms governing the activity of the ChREBP isoforms, including PTMs, partners/cofactors as well as novel metabolic pathways regulated by ChREBP in key metabolic tissues, by discussing phenotypes associated with tissue-specific deletion of ChREBP in knockout mice.
Collapse
Affiliation(s)
| | - Catherine Postic
- Université de Paris, Institut Cochin, CNRS, INSERM, Paris, France
| |
Collapse
|
46
|
Wu S, Näär AM. A lipid-free and insulin-supplemented medium supports De Novo fatty acid synthesis gene activation in melanoma cells. PLoS One 2019; 14:e0215022. [PMID: 30970006 PMCID: PMC6457551 DOI: 10.1371/journal.pone.0215022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/25/2019] [Indexed: 12/23/2022] Open
Abstract
While investigating the role played by de novo lipid (DNL) biosynthesis in cancer cells, we sought a medium condition that would support cell proliferation without providing any serum lipids. Here we report that a defined serum free cell culture medium condition containing insulin, transferrin and selenium (ITS) supports controlled study of transcriptional regulation of de novo fatty acid (DNFA) production and de novo cholesterol synthesis (DNCS) in melanoma cell lines. This lipid-free ITS medium is able to support continuous proliferation of several melanoma cell lines that utilize DNL to support their lipid requirements. We show that the ITS medium stimulates gene transcription in support of both DNFA and DNCS, specifically mediated by SREBP1/2 in melanoma cells. We further found that the ITS medium promoted SREBP1 nuclear localization and occupancy on DNFA gene promoters. Our data show clear utility of this serum and lipid-free medium for melanoma cancer cell culture and lipid-related areas of investigation.
Collapse
Affiliation(s)
- Su Wu
- Massachusetts General Hospital Center for Cancer Research, Charlestown, Massachusetts, United States of America
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (SW); (AMN)
| | - Anders M. Näär
- Massachusetts General Hospital Center for Cancer Research, Charlestown, Massachusetts, United States of America
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (SW); (AMN)
| |
Collapse
|
47
|
Lee J, Ridgway ND. Substrate channeling in the glycerol-3-phosphate pathway regulates the synthesis, storage and secretion of glycerolipids. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1865:158438. [PMID: 30959116 DOI: 10.1016/j.bbalip.2019.03.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 01/16/2023]
Abstract
The successive acylation of glycerol-3-phosphate (G3P) by glycerol-3-phosphate acyltransferases and acylglycerol-3-phosphate acyltransferases produces phosphatidic acid (PA), a precursor for CDP-diacylglycerol-dependent phospholipid synthesis. PA is further dephosphorylated by LIPINs to produce diacylglycerol (DG), a substrate for the synthesis of triglyceride (TG) by DG acyltransferases and a precursor for phospholipid synthesis via the CDP-choline and CDP-ethanolamine (Kennedy) pathways. The channeling of fatty acids into TG for storage in lipid droplets and secretion in lipoproteins or phospholipids for membrane biogenesis is dependent on isoform expression, activity and localization of G3P pathway enzymes, as well as dietary and hormonal and tissue-specific factors. Here, we review the mechanisms that control partitioning of substrates into lipid products of the G3P pathway.
Collapse
Affiliation(s)
- Jonghwa Lee
- Atlantic Research Center, Depts. of Pediatrics and Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS, Canada
| | - Neale D Ridgway
- Atlantic Research Center, Depts. of Pediatrics and Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
48
|
Soya protein β-conglycinin ameliorates fatty liver and obesity in diet-induced obese mice through the down-regulation of PPARγ. Br J Nutr 2019; 119:1220-1232. [PMID: 29770757 DOI: 10.1017/s0007114518000739] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Diets high in fat can result in obesity and non-alcoholic fatty liver disease (NAFLD). The improvement of obesity and NAFLD is an important issue. β-Conglycinin, one of the soya proteins, is known to prevent hyperlipidaemia, obesity and NAFLD. Therefore, we aimed to investigate the effects of β-conglycinin on the improvement of obesity and NAFLD in high-fat (HF) diet-induced obese (DIO) mice and clarify the mechanism underlying these effects in liver and white adipose tissue (WAT). DIO male ddY mice were divided into six groups: HF, medium-fat (MF) and low-fat (LF) groups fed casein, and HF, MF and LF groups in all of which the casein was replaced by β-conglycinin. A period of 5 weeks later, the β-conglycinin-supplemented group resulted in lower body weight, relative weight of subcutaneous WAT, and hepatic TAG content (P=0·001). Furthermore, β-conglycinin suppressed the hepatic expression of Pparγ2 in the HF dietary group, sterol regulatory element-binding protein-1c and the target genes. The expressions of inflammation-related genes were significantly low in the epididymal and subcutaneous WAT from the mice fed β-conglycinin compared with those fed casein in the HF dietary group. Moreover, the expressions of Pparγ1 and Pparγ2 mRNA were suppressed in subcutaneous WAT in the HF dietary group but not in epididymal WAT. The concentrations of insulin and leptin were low in the serum of the mice fed β-conglycinin. In conclusion, β-conglycinin effectively improved obesity and NAFLD in DIO mice, and it appears to be a promising dietary protein for the amelioration of NAFLD and obesity.
Collapse
|
49
|
Xu HF, Luo J, Zhang XY, Li J, Bionaz M. Activation of liver X receptor promotes fatty acid synthesis in goat mammary epithelial cells via modulation of SREBP1 expression. J Dairy Sci 2019; 102:3544-3555. [PMID: 30738675 DOI: 10.3168/jds.2018-15538] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 12/12/2018] [Indexed: 01/04/2023]
Abstract
In bovine mammary tissue and cells, liver X receptor (LXR) regulates lipid synthesis mainly via transactivation of the transcription factor sterol regulatory element binding protein 1 (SREBP1). In the present work, we investigated the role of LXR in controlling lipid synthesis via transactivation of SREBP1 in goat primary mammary cells (GMEC). The GMEC were treated with a synthetic agonist of LXR, T0901317, and transactivation and transcription of SREBP1, expression of lipogenic genes, and fatty acid profiling and triacylglycerol (TAG) content of the cells were measured. A mild increase in the mRNA expression level of LXRα (NR1H3) was observed following treatment with different concentrations of T0901317, and a dose-dependent increase in mRNA and transactivation of SREBP1 was detected. Activation of LXR resulted in a significant increase in the mRNA expression of most of the measured genes related to de novo synthesis, desaturation, and transport of fatty acids; TAG synthesis; and transcription regulators. Compared with the control, total content of cellular TAG increased by more than 20% with T0901317 treatment. Furthermore, addition of T0901317 increased the proportion of unsaturated fatty acids (e.g., C16:1, C18:1, C20:1, and C22:1), and decreased the proportion of saturated fatty acids (e.g., C16:0, C18:0, C20:0, and C22:0). These results provide evidence that LXR regulates the expression and activity of SREBP1. Our results indicated that LXR participate in regulating the transcription of genes involved in milk fat synthesis in GMEC in an SREBP1-dependent fashion.
Collapse
Affiliation(s)
- H F Xu
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, P. R. China
| | - J Luo
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China.
| | - X Y Zhang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - J Li
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, Henan, 450046, P. R. China
| | - M Bionaz
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis 97331.
| |
Collapse
|
50
|
Li H, Liu Y, Liu C, Luo L, Yao Y, Li F, Yin L, Xu L, Tong Q, Huang C, Fan S. Notoginsenoside Fe suppresses diet induced obesity and activates paraventricular hypothalamic neurons. RSC Adv 2019; 9:1290-1298. [PMID: 35518019 PMCID: PMC9059641 DOI: 10.1039/c8ra07842d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 12/24/2018] [Indexed: 01/13/2023] Open
Abstract
Obesity has become a major public health challenge worldwide. Energy imbalance between calorie acquisition and consumption is the fundamental cause of obesity. Notoginsenoside Fe is a naturally occurring compound in Panax notoginseng, a herb used in the treatment of cardiovascular diseases in traditional Chinese medicine. Here, we evaluated the effect of notoginsenoside Fe on obesity development induced by high-fat diet in C57BL/6 mice. Our results demonstrated that notoginsenoside Fe decreased food intake and body weight, as well as protected liver structure integrity and normal function. Metabolic cage analysis showed that notoginsenoside Fe also promoted resting metabolic rate. In addition, intracerebroventricular (i.c.v) injection of notoginsenoside Fe induced C-Fos expression in the paraventricular nucleus (PVH) but not the arcuate nucleus (ARC) of the hypothalamus. These results suggest that Fe may reduce body weight through the activation of energy-sensing neurons in the hypothalamus. Notoginsenoside Fe, a naturally occurring compound in Panax notoginseng, significantly reduces body weight, promotes metabolic rate, and suppresses food intake through activating C-Fos expression in PVH in high-fat diet induced obese mice.![]()
Collapse
|