51
|
Witka BZ, Oktaviani DJ, Marcellino M, Barliana MI, Abdulah R. Type 2 Diabetes-Associated Genetic Polymorphisms as Potential Disease Predictors. Diabetes Metab Syndr Obes 2019; 12:2689-2706. [PMID: 31908510 PMCID: PMC6927489 DOI: 10.2147/dmso.s230061] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/19/2019] [Indexed: 12/18/2022] Open
Abstract
Diabetes is a major cause of mortality worldwide. There are several types of diabetes, with type 2 diabetes mellitus (T2DM) being the most common. Many factors, including environmental and genetic factors, are involved in the etiology of the disease. Numerous studies have reported the role of genetic polymorphisms in the initiation and development of T2DM. While genome-wide association studies have identified around more than 200 susceptibility loci, it remains unclear whether these loci are correlated with the pathophysiology of the disease. The present review aimed to elucidate the potential genetic mechanisms underlying T2DM. We found that some genetic polymorphisms were related to T2DM, either in the form of single-nucleotide polymorphisms or direct amino acid changes in proteins. These polymorphisms are potential predictors for the management of T2DM.
Collapse
Affiliation(s)
- Beska Z Witka
- Departement of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
| | - Dede J Oktaviani
- Departement of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
| | - Marcellino Marcellino
- Departement of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
| | - Melisa I Barliana
- Departement of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Jatinangor, Indonesia
- Correspondence: Melisa I Barliana Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung Sumedang KM. 21, Jatinangor45363, Indonesia Email
| | - Rizky Abdulah
- Departement of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Jatinangor, Indonesia
| |
Collapse
|
52
|
Wang Z, Dong C. Gluconeogenesis in Cancer: Function and Regulation of PEPCK, FBPase, and G6Pase. Trends Cancer 2018; 5:30-45. [PMID: 30616754 DOI: 10.1016/j.trecan.2018.11.003] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 11/11/2018] [Accepted: 11/12/2018] [Indexed: 01/19/2023]
Abstract
Cancer cells display a high rate of glycolysis in the presence of oxygen to promote proliferation. Gluconeogenesis, the reverse pathway of glycolysis, can antagonize aerobic glycolysis in cancer via three key enzymes - phosphoenolpyruvate carboxykinase (PEPCK), fructose-1,6-bisphosphatase (FBPase), and glucose-6-phosphatase (G6Pase). Recent studies have revealed that, in addition to metabolic regulation, these enzymes also play a role in signaling, proliferation, and the cancer stem cell (CSC) tumor phenotype. Multifaceted regulation of PEPCK, FBPase, and G6Pase through transcription, epigenetics, post-translational modification, and enzymatic activity is observed in different cancers. We review here the function and regulation of key gluconeogenic enzymes and new therapeutic opportunities.
Collapse
Affiliation(s)
- Zhanyu Wang
- Department of Pathology and Pathophysiology, and Department of Surgical Oncology (Breast Center) of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Chenfang Dong
- Department of Pathology and Pathophysiology, and Department of Surgical Oncology (Breast Center) of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
53
|
Puppala S, Li C, Glenn JP, Saxena R, Gawrieh S, Quinn A, Palarczyk J, Dick EJ, Nathanielsz PW, Cox LA. Primate fetal hepatic responses to maternal obesity: epigenetic signalling pathways and lipid accumulation. J Physiol 2018; 596:5823-5837. [PMID: 29516496 PMCID: PMC6265567 DOI: 10.1113/jp275422] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 02/01/2018] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS Maternal obesity (MO) and exposure to a high-fat, high-simple-carbohydrate diet during pregnancy predisposes offspring to obesity, metabolic and cardiovascular disorders in later life. Underlying molecular pathways and potential epigenetic factors that are dysregulated in MO were identified using unbiased transcriptomic methods. There was increased lipid accumulation and severe steatosis in the MO baboon fetal liver suggesting that these offspring are on an early trajectory of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis. ABSTRACT Maternal obesity (MO) increases offspring cardiometabolic disease risk. Altered fetal liver development in response to the challenge of MO has metabolic consequences underlying adverse offspring life-course health outcomes. Little is known about the molecular pathways and potential epigenetic changes regulating primate fetal liver responses to MO. We hypothesized that MO would induce fetal baboon liver epigenetic changes resulting in dysregulation of key metabolic pathways that impact lipid metabolism. MO was induced prior to pregnancy by a high-fat, high-fructose diet. Unbiased gene and microRNA (small RNA Seq) abundance analyses were performed on fetal baboon livers at 0.9 gestation and subjected to pathway analyses to identify fetal liver molecular responses to MO. Fetal baboon liver lipid and glycogen content were quantified by the Computer Assisted Stereology Toolbox. In response to MO, fetal livers revealed dysregulation of TCA cycle, proteasome, oxidative phosphorylation, glycolysis and Wnt/β-catenin signalling pathways together with marked lipid accumulation supporting our hypothesis that multiple pathway dysregulation detrimentally impacts lipid management. This is the first study of MO programming of the non-human primate fetal liver using unbiased transcriptome analysis to detect changes in hepatic gene expression levels and identify potential microRNA epigenetic regulators of metabolic disruption.
Collapse
Affiliation(s)
- Sobha Puppala
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest BaptistMedical CenterWinston‐SalemNCUSA
| | - Cun Li
- Department of Animal ScienceUniversity of WyomingLaramieWYUSA
| | - Jeremy P. Glenn
- Department of GeneticsTexas Biomedical Research InstituteSan AntonioTXUSA
| | - Romil Saxena
- Department of Pathology, Indiana University School of MedicineIndianapolisINUSA
| | - Samer Gawrieh
- Division of Gastroenterology and HepatologyIndiana University School of MedicineIndianapolisINUSA
| | - Amy Quinn
- Department of Pediatrics, Division of NeonatologyUniversity of Texas Health Science CenterSan AntonioTXUSA
| | - Jennifer Palarczyk
- Department of Pediatrics, Division of NeonatologyUniversity of Texas Health Science CenterSan AntonioTXUSA
| | - Edward J. Dick
- Southwest National Primate Research CenterTexas Biomedical Research InstituteSan AntonioTXUSA
| | - Peter W. Nathanielsz
- Department of Animal ScienceUniversity of WyomingLaramieWYUSA
- Department of GeneticsTexas Biomedical Research InstituteSan AntonioTXUSA
| | - Laura A. Cox
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest BaptistMedical CenterWinston‐SalemNCUSA
- Southwest National Primate Research CenterTexas Biomedical Research InstituteSan AntonioTXUSA
| |
Collapse
|
54
|
Fritsche L, Sarief M, Wagner R, Stefan N, Lehmann R, Häring HU, Grallert H, Fritsche A, Lechner A. Genetic variation in TCF7L2 rs7903146 and history of GDM negatively and independently impact on diabetes-associated metabolic traits. Diabetes Res Clin Pract 2018; 146:251-257. [PMID: 30419301 DOI: 10.1016/j.diabres.2018.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/02/2018] [Accepted: 11/02/2018] [Indexed: 12/22/2022]
Abstract
AIMS Gestational diabetes (GDM) is recognized as a major risk factor for the development of type 2 diabetes (T2DM) later in life. Risk allele carriers at TCF7L2 rs7903146 have increased susceptibility for both GDM and T2DM. We hypothesized that carrying TCF7L2 risk alleles would further aggravate the negative impact of a positive history for GDM on metabolic traits related to T2DM later in life. METHODS 210 women with a confirmed history of gestational diabetes and 810 controls without evidence for GDM underwent standardized 75 g oral glucose tolerance tests (OGTT). Liver fat was quantified in a subset of subjects (n = 444) using magnetic resonance spectroscopy. RESULTS 504 women were homozygous or heterozygous risk allele carriers. The risk allele carriers had a higher risk for GDM (p = 0.0076, OR 1.52, 95% CI 1.11-2.06). Multivariable regression analysis demonstrated that both a history of GDM, or carrying a TCF7L2 risk allele resulted in lower insulin secretion, impaired proinsulin processing and higher fasting and 2-hour glucose levels. Liver fat content was not associated with either a history of GDM or a TCF7L2 risk genotype. There was no significant interaction (all p > 0.05) between history of GDM and TCF7L2 risk alleles on all diabetes-associated metabolic traits tested. CONCLUSION The TCF7L2 rs7903146 polymorphism is a risk factor for gestational diabetes. However, the additional presence of TCF7L2 rs7903146 risk alleles does not further aggravate the negative impact of a history of gestational diabetes on metabolic traits related to T2DM.
Collapse
Affiliation(s)
- Louise Fritsche
- Division of Endocrinology, Diabetology, Nephrology, Angiology and Clinical Chemistry, Eberhardt Karls University, Tuebingen, Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tuebingen (IDM), Tuebingen, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Mirjam Sarief
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Diabetes Research Group, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany; Clinical Cooperation Group Type 2 Diabetes, Helmholtz Center Munich, Neuherberg, Germany
| | - Robert Wagner
- Division of Endocrinology, Diabetology, Nephrology, Angiology and Clinical Chemistry, Eberhardt Karls University, Tuebingen, Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tuebingen (IDM), Tuebingen, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Norbert Stefan
- Division of Endocrinology, Diabetology, Nephrology, Angiology and Clinical Chemistry, Eberhardt Karls University, Tuebingen, Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tuebingen (IDM), Tuebingen, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Rainer Lehmann
- Division of Endocrinology, Diabetology, Nephrology, Angiology and Clinical Chemistry, Eberhardt Karls University, Tuebingen, Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tuebingen (IDM), Tuebingen, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Hans-Ulrich Häring
- Division of Endocrinology, Diabetology, Nephrology, Angiology and Clinical Chemistry, Eberhardt Karls University, Tuebingen, Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tuebingen (IDM), Tuebingen, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Harald Grallert
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Diabetes Research Group, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany; Research Unit of Molecular Epidemiology, Institute for Epidemiology II, Helmholtz Center Munich, Neuherberg, Germany
| | - Andreas Fritsche
- Division of Endocrinology, Diabetology, Nephrology, Angiology and Clinical Chemistry, Eberhardt Karls University, Tuebingen, Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tuebingen (IDM), Tuebingen, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Andreas Lechner
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Diabetes Research Group, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany; Clinical Cooperation Group Type 2 Diabetes, Helmholtz Center Munich, Neuherberg, Germany
| |
Collapse
|
55
|
Wu K, Wang L, Chen Y, Pirooznia M, Singh K, Wälde S, Kehlenbach RH, Scott I, Gucek M, Sack MN. GCN5L1 interacts with αTAT1 and RanBP2 to regulate hepatic α-tubulin acetylation and lysosome trafficking. J Cell Sci 2018; 131:jcs.221036. [PMID: 30333138 DOI: 10.1242/jcs.221036] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 10/04/2018] [Indexed: 01/07/2023] Open
Abstract
Although GCN5L1 (also known as BLOC1S1) facilitates mitochondrial protein acetylation and controls endosomal-lysosomal trafficking, the mechanisms underpinning these disparate effects are unclear. As microtubule acetylation modulates endosome-lysosome trafficking, we reasoned that exploring the role of GCN5L1 in this biology may enhance our understanding of GCN5L1-mediated protein acetylation. We show that α-tubulin acetylation is reduced in GCN5L1-knockout hepatocytes and restored by GCN5L1 reconstitution. Furthermore, GCN5L1 binds to the α-tubulin acetyltransferase αTAT1, and GCN5L1-mediated α-tubulin acetylation is dependent on αTAT1. Given that cytosolic GCN5L1 has been identified as a component of numerous multiprotein complexes, we explored whether novel interacting partners contribute to this regulation. We identify RanBP2 as a novel interacting partner of GCN5L1 and αTAT1. Genetic silencing of RanBP2 phenocopies GCN5L1 depletion by reducing α-tubulin acetylation, and we find that RanBP2 possesses a tubulin-binding domain, which recruits GCN5L1 to α-tubulin. Finally, we find that genetic depletion of GCN5L1 promotes perinuclear lysosome accumulation and histone deacetylase inhibition partially restores lysosomal positioning. We conclude that the interactions of GCN5L1, RanBP2 and αTAT1 function in concert to control α-tubulin acetylation and may contribute towards the regulation of cellular lysosome positioning. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Kaiyuan Wu
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lingdi Wang
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yong Chen
- Proteomics Core, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Mehdi Pirooznia
- Bioinformatics and Computational Biology Core, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Komudi Singh
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sarah Wälde
- Department of Molecular Biology, Faculty of Medicine, Georg-August-University Göttingen, 37073 Göttingen, Germany
| | - Ralph H Kehlenbach
- Department of Molecular Biology, Faculty of Medicine, Georg-August-University Göttingen, 37073 Göttingen, Germany
| | - Iain Scott
- Cardiology Division, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA
| | - Marjan Gucek
- Proteomics Core, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Michael N Sack
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
56
|
Chen D, Gong Y, Xu L, Zhou M, Li J, Song J. Bidirectional regulation of osteogenic differentiation by the FOXO subfamily of Forkhead transcription factors in mammalian MSCs. Cell Prolif 2018; 52:e12540. [PMID: 30397974 PMCID: PMC6496202 DOI: 10.1111/cpr.12540] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 08/09/2018] [Accepted: 09/02/2018] [Indexed: 12/23/2022] Open
Abstract
Through loss‐ and gain‐of‐function experiments in knockout and transgenic mice, Forkhead box O (FOXO) family transcription factors have been demonstrated to play essential roles in many biological processes, including cellular proliferation, apoptosis and differentiation. Osteogenic differentiation from mesenchymal stem cells (MSCs) into osteoblasts is a well‐organized process that is carefully guided and characterized by various factors, such as runt‐related transcription factor 2 (Runx2), β‐catenin, osteocalcin (OCN), alkaline phosphatase (ALP) and activating transcription factor 4 (ATF4). Accumulating evidence suggests multiple interactions among FOXO members and the differentiation regulatory factors listed above, resulting in an enhancement or inhibition of osteogenesis in different stages of osteogenic differentiation. To systematically and integrally understand the role of FOXOs in osteogenic differentiation and explain the contrary phenomena observed in vitro and in vivo, we herein summarized FOXO‐interacting differentiation regulatory genes/factors and following alterations in differentiation. The underlying mechanism was further discussed on the basis of binding types, sites, phases and the consequent downstream transcriptional alterations of interactions among FOXOs and differentiation regulatory factors. Interestingly, a bidirectional effect of FOXOs on balancing osteogenic differentiation was discovered in MSCs. Moreover, FOXO factors are reported to be activated or suppressed by several context‐dependent signalling inputs during differentiation, and the underlying molecular basis may offer new drug development targets for treatments of bone formation defect diseases.
Collapse
Affiliation(s)
- Duanjing Chen
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Yuanyuan Gong
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Ling Xu
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Mengjiao Zhou
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jie Li
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jinlin Song
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| |
Collapse
|
57
|
Langlet F, Tarbier M, Haeusler RA, Camastra S, Ferrannini E, Friedländer MR, Accili D. microRNA-205-5p is a modulator of insulin sensitivity that inhibits FOXO function. Mol Metab 2018; 17:49-60. [PMID: 30174230 PMCID: PMC6197154 DOI: 10.1016/j.molmet.2018.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 08/07/2018] [Indexed: 12/20/2022] Open
Abstract
Objectives Hepatic insulin resistance is a hallmark of type 2 diabetes and obesity. Insulin receptor signaling through AKT and FOXO has important metabolic effects that have traditionally been ascribed to regulation of gene expression. However, whether all the metabolic effects of FOXO arise from its regulation of protein-encoding mRNAs is unknown. Methods To address this question, we obtained expression profiles of FOXO-regulated murine hepatic microRNAs (miRNAs) during fasting and refeeding using mice lacking Foxo1, 3a, and 4 in liver (L-Foxo1,3a, 4). Results Out of 439 miRNA analyzed, 175 were differentially expressed in Foxo knockouts. Their functions were associated with insulin, Wnt, Mapk signaling, and aging. Among them, we report a striking increase of miR-205-5p expression in L-Foxo1,3a,4 knockouts, as well as in obese mice. We show that miR-205-5p gain-of-function increases AKT phosphorylation and decreases SHIP2 in primary hepatocytes, resulting in FOXO inhibition. This results in decreased hepatocyte glucose production. Consistent with these observations, miR-205-5p gain-of-function in mice lowered glucose levels and improved pyruvate tolerance. Conclusions These findings reveal a homeostatic miRNA loop regulating insulin signaling, with potential implications for in vivo glucose metabolism. A comprehensive analysis of Foxo-dependent miRNA. miRNAs recapitulate the transcriptional effects of Foxo on insulin signaling. Foxo regulates miRNA transcription during the fasting/refeeding transition. miR205 regulates insulin sensitivity through a homeostatic loop with Foxo.
Collapse
Affiliation(s)
- Fanny Langlet
- Naomi Berrie Diabetes Center and Departments of Medicine, Columbia University, New York, 10032, USA
| | - Marcel Tarbier
- Science for Life Laboratory, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 17121, Stockholm, Sweden
| | - Rebecca A Haeusler
- Naomi Berrie Diabetes Center and Departments of Pathology and Cell Biology, Columbia University, New York, 10032, USA
| | - Stefania Camastra
- Department of Clinical and Experimental Medicine, University of Pisa School of Medicine, Pisa, Italy
| | - Eleuterio Ferrannini
- Department of Clinical and Experimental Medicine, University of Pisa School of Medicine, Pisa, Italy; CNR Institute of Clinical Physiology, Pisa, Italy
| | - Marc R Friedländer
- Science for Life Laboratory, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 17121, Stockholm, Sweden
| | - Domenico Accili
- Naomi Berrie Diabetes Center and Departments of Medicine, Columbia University, New York, 10032, USA.
| |
Collapse
|
58
|
Leibing T, Géraud C, Augustin I, Boutros M, Augustin HG, Okun JG, Langhans C, Zierow J, Wohlfeil SA, Olsavszky V, Schledzewski K, Goerdt S, Koch P. Angiocrine Wnt signaling controls liver growth and metabolic maturation in mice. Hepatology 2018; 68:707-722. [PMID: 29059455 PMCID: PMC6099291 DOI: 10.1002/hep.29613] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 09/08/2017] [Accepted: 10/19/2017] [Indexed: 12/21/2022]
Abstract
UNLABELLED Postnatal liver development is characterized by hepatocyte growth, proliferation, and functional maturation. Notably, canonical Wnt signaling in hepatocytes has been identified as an important regulator of final adult liver size and metabolic liver zonation. The cellular origin of Wnt ligands responsible for homeostatic liver/body weight ratio (LW/BW) remained unclear, which was also attributable to a lack of suitable endothelial Cre driver mice. To comprehensively analyze the effects of hepatic angiocrine Wnt signaling on liver development and metabolic functions, we used endothelial subtype-specific Stab2-Cre driver mice to delete Wls from hepatic endothelial cells (HECs). The resultant Stab2-Cretg/wt ;Wlsfl/fl (Wls-HECKO) mice were viable, but showed a significantly reduced LW/BW. Specifically, ablation of angiocrine Wnt signaling impaired metabolic zonation in the liver, as shown by loss of pericentral, β-catenin-dependent target genes such as glutamine synthase (Glul), RhBg, Axin2, and cytochrome P450 2E1, as well as by extended expression of periportal genes such as arginase 1. Furthermore, endothelial subtype-specific expression of a c-terminally YFP-tagged Wls fusion protein in Wls-HECKO mice (Stab2-Cretg/wt ;Wlsfl/fl ;Rosa26:Wls-YFPfl/wt [Wls-rescue]) restored metabolic liver zonation. Interestingly, lipid metabolism was altered in Wls-HECKO mice exhibiting significantly reduced plasma cholesterol levels, while maintaining normal plasma triglyceride and blood glucose concentrations. On the contrary, zonal expression of Endomucin, LYVE1, and other markers of HEC heterogeneity were not altered in Wls-HECKO livers. CONCLUSION Angiocrine Wnt signaling controls liver growth as well as development of metabolic liver zonation in mice, whereas intrahepatic HEC zonation is not affected. (Hepatology 2017).
Collapse
Affiliation(s)
- Thomas Leibing
- Department of Dermatology, Venereology, and AllergologyUniversity Medical Center and Medical Faculty Mannheim, Heidelberg University, and Center of Excellence in DermatologyMannheimGermany
| | - Cyrill Géraud
- Department of Dermatology, Venereology, and AllergologyUniversity Medical Center and Medical Faculty Mannheim, Heidelberg University, and Center of Excellence in DermatologyMannheimGermany
| | - Iris Augustin
- German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics and Heidelberg University, Faculty of Medicine Mannheim, Department of Cell and Molecular BiologyHeidelbergGermany,Molecular Cell Biology and Plant Cell TechnologyUniversity of Applied Sciences Weihenstephan‐TriesdorfFreisingGermany
| | - Michael Boutros
- German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics and Heidelberg University, Faculty of Medicine Mannheim, Department of Cell and Molecular BiologyHeidelbergGermany
| | - Hellmut G. Augustin
- Division of Vascular Oncology and Metastasis (DKFZ‐ZMBH Alliance)DKFZHeidelbergGermany,Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty MannheimHeidelberg UniversityMannheimGermany
| | - Jürgen G. Okun
- Department of General Pediatrics, Division of Inherited Metabolic DiseasesUniversity Children's HospitalHeidelbergGermany
| | - Claus‐Dieter Langhans
- Department of General Pediatrics, Division of Inherited Metabolic DiseasesUniversity Children's HospitalHeidelbergGermany
| | - Johanna Zierow
- Department of Dermatology, Venereology, and AllergologyUniversity Medical Center and Medical Faculty Mannheim, Heidelberg University, and Center of Excellence in DermatologyMannheimGermany
| | - Sebastian A. Wohlfeil
- Department of Dermatology, Venereology, and AllergologyUniversity Medical Center and Medical Faculty Mannheim, Heidelberg University, and Center of Excellence in DermatologyMannheimGermany
| | - Victor Olsavszky
- Department of Dermatology, Venereology, and AllergologyUniversity Medical Center and Medical Faculty Mannheim, Heidelberg University, and Center of Excellence in DermatologyMannheimGermany
| | - Kai Schledzewski
- Department of Dermatology, Venereology, and AllergologyUniversity Medical Center and Medical Faculty Mannheim, Heidelberg University, and Center of Excellence in DermatologyMannheimGermany
| | - Sergij Goerdt
- Department of Dermatology, Venereology, and AllergologyUniversity Medical Center and Medical Faculty Mannheim, Heidelberg University, and Center of Excellence in DermatologyMannheimGermany,European Center for AngioscienceMedical Faculty Mannheim, University of HeidelbergMannheimGermany
| | - Philipp‐Sebastian Koch
- Department of Dermatology, Venereology, and AllergologyUniversity Medical Center and Medical Faculty Mannheim, Heidelberg University, and Center of Excellence in DermatologyMannheimGermany
| |
Collapse
|
59
|
Huang HH, Yeh C, Chen JC, Lee TH, Chen SC, Lee WJ, Chen CY. Does bariatric surgery influence plasma levels of fetuin-A and leukocyte cell-derived chemotaxin-2 in patients with type 2 diabetes mellitus? PeerJ 2018; 6:e4884. [PMID: 29910974 PMCID: PMC6003398 DOI: 10.7717/peerj.4884] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 05/11/2018] [Indexed: 12/11/2022] Open
Abstract
Background Fetuin-A and leukocyte cell-derived chemotaxin-2 (LECT-2) are liver-derived proteins. Fetuin-A is an independent risk factor for type 2 diabetes (T2D) and obese patients with T2D have higher plasma fetuin-A levels than those without T2D. LECT-2 has positive correlation with the severity of both obesity and insulin resistance. The changes in plasma fetuin-A are not consistent after bariatric surgery and no studies have investigated the changes in LECT-2 on the obese patients with T2D after bariatric surgery. Methods Overall, 18 patients undergoing gastric bypass (GB) and 16 patients undergoing sleeve gastrectomy (SG) were enrolled. The fasting plasma fetuin-A and LECT-2 levels were measured at baseline, one week, three months, and one year after surgery. Results Both the GB and SG groups significantly decreased the body mass index (BMI), waist-to-hip ratio, a body shape index; the triglyceride, fasting blood sugar (FBS), hemoglobin A1c, C-peptide levels; and homeostatic model assessment (HOMA-IR) one year after surgery. The SG group showed a decreasing trend in plasma fetuin-A levels one year after SG surgery. There are no significant changes in LECT-2 one year after either GB or SG. Fetuin-A had a near significant negative relationship with insulin (P = 0.056) and HOMA-IR (P = 0.050) in the SG group. Changes in fetuin-A had a significant positive relationship with changes in BMI (P = 0.031) and waist-to-hip ratio (P = 0.031) in the GB group and had a near significant positive correlation with FBS (P = 0.051) in the SG group. Discussion Neither GB nor SG modifies plasma levels of plasma fetuin-A or LECT-2 in T2D patients after surgery. The changes in plasma fetuin-A have a positive correlation with those of the BMI and waist-to-hip ratio 12 months after GB.
Collapse
Affiliation(s)
- Hsien-Hao Huang
- Department of Emergency Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Emergency and Critical Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Chun Yeh
- Division of Gastroenterology, Department of Internal Medicine, Cheng-Hsin General Hospital, Taipei, Taiwan.,Department of Internal Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Jung-Chien Chen
- Department of Surgery, Min-Sheng General Hospital, Taoyuan, Taiwan
| | - Tzong-Hsi Lee
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan.,Department of Nursing, Oriental Institute of Technology, New Taipei, Taiwan
| | - Shu-Chun Chen
- Department of Nursing, Min-Sheng General Hospital, Taoyuan, Taiwan.,Taiwan Society for Metabolic and Bariatric Surgery, Taoyuan, Taiwan
| | - Wei-Jei Lee
- Department of Surgery, Min-Sheng General Hospital, Taoyuan, Taiwan.,Taiwan Society for Metabolic and Bariatric Surgery, Taoyuan, Taiwan
| | - Chih-Yen Chen
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Bariatric and Metabolic Surgery Center, Taipei Veterans General Hospital, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Taiwan Association for the Study of Small Intestinal Diseases, Guishan, Taiwan.,Chinese Taipei Society for the Study of Obesity, Taipei, Taiwan
| |
Collapse
|
60
|
Salvianolic acid A alleviates chronic ethanol-induced liver injury via promotion of β-catenin nuclear accumulation by restoring SIRT1 in rats. Toxicol Appl Pharmacol 2018; 350:21-31. [PMID: 29729281 DOI: 10.1016/j.taap.2018.04.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 04/27/2018] [Accepted: 04/29/2018] [Indexed: 02/07/2023]
Abstract
In recent years, alcoholic liver disease (ALD) has emerged as a growing public health problem worldwide. β-catenin plays an important role in the growth, development, regeneration and metabolic activity of the liver. Salvianolic acid A (SalA) is a water-soluble component from the root extract of Salvia miltiorrhiza Bunge, and its effect on ALD has not yet been investigated. This study aimed to investigate the effect of SalA on chronic alcohol-induced liver injury and to explore the role of SIRT1-mediated β-catenin deacetylation in such an effect. In this study, SalA treatment significantly alleviated the accumulation of lipid droplets and reduced the plasma alanine aminotransferase (ALT), aspartate aminotransferase (AST), total cholesterol (TC), triglyceride (TG), alcohol and ammonia levels in rats. SalA enhanced ethanol and ammonia metabolism and maintained mitochondrial homeostasis. Moreover, SalA restored the activity of the major ethanol-metabolizing enzymes and oxidative stress functions in the liver. Importantly, we found that SalA treatment effectively inhibited the ethanol-mediated decrease in nuclear β-catenin by upregulating SIRT1 in the liver. SIRT1 then deacetylated β-catenin to promote its accumulation in the nucleus, thereby preventing alcohol-induced liver injury. The results demonstrate that the SIRT1/β-catenin pathway is a key therapeutic target in liver injury caused by chronic alcohol exposure and that SalA protects against alcohol-induced liver injury via the SIRT1-mediated deacetylation of β-catenin.
Collapse
|
61
|
Population based and animal study on the effects of Schistosoma japonicum infection in the regulation of host glucose homeostasis. Acta Trop 2018; 180:33-41. [PMID: 29309743 DOI: 10.1016/j.actatropica.2018.01.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 11/19/2017] [Accepted: 01/05/2018] [Indexed: 01/17/2023]
Abstract
Although parasitic infection affects the glucose homeostasis of mice, only few studies have integrated epidemiological and animal data to determine the effect of Schistosoma japonicum infection on mice metabolism. The current study assessed the effects of S. japonicum infection on blood glucose and other metabolic parameters in both patients and animal models of chronic schistomiasis. A total of 2183 patients with chronic schistosomiasis and age- and gender-matched individuals without schistosomiasis (n = 1798) were enrolled in this study. Fasting blood glucose and other metabolic parameters, including body mass index (BMI) and serum triglyceride and total cholesterol, were compared between the two groups. Mice infected with S. japonicum were used to test the effects of the parasite on glucose tolerance. We found that chronic schistosomiasis patients had significantly lower BMI and fasting blood glucose, serum triglyceride, and total cholesterol levels than non-schistosomiasis individuals. In the animal studies, both bisexual and unisexual S. japonicum infection improved glucose tolerance in wild-type mice. Additionally, S. japonicum-infected ob/ob mice, a model that spontaneously develops obesity and diabetes, also had decreased body weight and improved glucose tolerance. We further observed that S. japonicum-infected mice had lower inflammatory gene expression in the visceral white adipose tissue than the control mice. Collectively, our results demonstrated that S. japonicum infection improved glucose tolerance and other metabolic parameters both in human and animals. Downregulated inflammatory gene expression due to S. japonicum infection might be among the mechanisms for the improved glucose tolerance.
Collapse
|
62
|
Qin Y, Chen M, Yang Y, Zhou XR, Shao SY, Wang DW, Yuan G. Liraglutide improves hepatic insulin resistance via the canonical Wnt signaling pathway. Mol Med Rep 2018; 17:7372-7380. [PMID: 29568881 DOI: 10.3892/mmr.2018.8737] [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] [Received: 03/07/2017] [Accepted: 11/23/2017] [Indexed: 11/06/2022] Open
Abstract
Liraglutide, a modified form of glucagon‑like peptide‑1 (GLP‑1), is used in the treatment of diabetes mellitus. However, the underlying mechanism by which liraglutide improves liver insulin resistance remains to be elucidated. The proto‑oncogene Wnt (Wnt) signaling pathway has been reported to be associated with glucose and lipid metabolism. Using in vivo and in vitro models of diabetes and insulin resistance, it was investigated whether the beneficial effects of liraglutide on liver glucose metabolism are mediated by the Wnt signaling pathway. The results of the present study demonstrate that body weight, fasting blood glucose, insulin levels and the homeostasis model assessment for insulin resistance were markedly decreased in db/db mice treated with liraglutide compared with control mice. Liraglutide also improved liver morphology and reduced the accumulation of lipid droplets. Furthermore, the expression of glucose-6-phosphatase and phosphoenolpyruvate carboxykinase was downregulated, whereas the expression of phosphorylated forkhead box O1, Wnt signaling pathway‑associated molecules, β‑catenin, transcription factor 7‑like 2 and phosphorylated glycogen synthase kinase-3β was upregulated in the liver of mice treated with liraglutide. In the in vitro study, increased gluconeogenesis and decreased glucose uptake rates were observed in insulin resistant hepatocytes; treatment with liraglutide significantly reversed this effect. Furthermore, transfection of insulin resistant hepatocytes with β‑catenin small interfering RNA attenuated the effects of liraglutide, suggesting that liraglutide improves insulin resistance via activating the β‑catenin/Wnt signaling pathway. The results of the present study suggest a novel mechanism underlying liraglutide‑mediated improvements in insulin resistance in the liver. The Wnt signaling pathway may be a potential therapeutic target for the treatment of altered hepatic physiology in insulin resistance.
Collapse
Affiliation(s)
- Yu Qin
- Department of Internal Medicine, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Min Chen
- Department of Geriatrics, The First Hospital of Jiangxia, Wuhan, Hubei 430030, P.R. China
| | - Yan Yang
- Department of Internal Medicine, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xin-Rong Zhou
- Department of Internal Medicine, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Shi-Ying Shao
- Department of Internal Medicine, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Dao-Wen Wang
- Department of Internal Medicine, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Gang Yuan
- Department of Internal Medicine, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
63
|
Nguyen LT, Reverter A, Cánovas A, Venus B, Anderson ST, Islas-Trejo A, Dias MM, Crawford NF, Lehnert SA, Medrano JF, Thomas MG, Moore SS, Fortes MRS. STAT6, PBX2, and PBRM1 Emerge as Predicted Regulators of 452 Differentially Expressed Genes Associated With Puberty in Brahman Heifers. Front Genet 2018; 9:87. [PMID: 29616079 PMCID: PMC5869259 DOI: 10.3389/fgene.2018.00087] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/02/2018] [Indexed: 12/17/2022] Open
Abstract
The liver plays a central role in metabolism and produces important hormones. Hepatic estrogen receptors and the release of insulin-like growth factor 1 (IGF1) are critical links between liver function and the reproductive system. However, the role of liver in pubertal development is not fully understood. To explore this question, we applied transcriptomic analyses to liver samples of pre- and post-pubertal Brahman heifers and identified differentially expressed (DE) genes and genes encoding transcription factors (TFs). Differential expression of genes suggests potential biological mechanisms and pathways linking liver function to puberty. The analyses identified 452 DE genes and 82 TF with significant contribution to differential gene expression by using a regulatory impact factor metric. Brain-derived neurotrophic factor was observed as the most down-regulated gene (P = 0.003) in post-pubertal heifers and we propose this gene influences pubertal development in Brahman heifers. Additionally, co-expression network analysis provided evidence for three TF as key regulators of liver function during pubertal development: the signal transducer and activator of transcription 6, PBX homeobox 2, and polybromo 1. Pathway enrichment analysis identified transforming growth factor-beta and Wnt signaling pathways as significant annotation terms for the list of DE genes and TF in the co-expression network. Molecular information regarding genes and pathways described in this work are important to further our understanding of puberty onset in Brahman heifers.
Collapse
Affiliation(s)
- Loan T Nguyen
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia.,Faculty of Biotechnology, Vietnam National University of Agriculture, Hanoi, Vietnam
| | - Antonio Reverter
- CSIRO Agriculture and Food, Queensland Bioscience Precinct, St. Lucia, QLD, Australia
| | - Angela Cánovas
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | - Bronwyn Venus
- Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St. Lucia, QLD, Australia
| | - Stephen T Anderson
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Alma Islas-Trejo
- Department of Animal Science, University of California, Davis, Davis, CA, United States
| | - Marina M Dias
- Departamento de Zootecnia, Faculdade de Ciências Agráìrias e Veterináìrias, Universidade Estadual Paulista Júlio de Mesquita Filho, São Paulo, Brazil
| | - Natalie F Crawford
- Department of Animal Science, Colorado State University, Fort Collins, CO, United States
| | - Sigrid A Lehnert
- CSIRO Agriculture and Food, Queensland Bioscience Precinct, St. Lucia, QLD, Australia
| | - Juan F Medrano
- Department of Animal Science, University of California, Davis, Davis, CA, United States
| | - Milt G Thomas
- Department of Animal Science, Colorado State University, Fort Collins, CO, United States
| | - Stephen S Moore
- Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St. Lucia, QLD, Australia
| | - Marina R S Fortes
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia.,Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St. Lucia, QLD, Australia
| |
Collapse
|
64
|
Rivas S, Gómez-Oro C, Antón IM, Wandosell F. Role of Akt Isoforms Controlling Cancer Stem Cell Survival, Phenotype and Self-Renewal. Biomedicines 2018. [PMID: 29518912 PMCID: PMC5874686 DOI: 10.3390/biomedicines6010029] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The cancer stem cell (CSC) hypothesis suggests that tumours are maintained by a subpopulation of cells with stem cell properties. Although the existence of CSCs was initially described in human leukaemia, less evidence exists for CSCs in solid tumours. Recently, a CD133+ cell subpopulation was isolated from human brain tumours exhibiting stem cell properties in vitro as well as the capacity to initiate tumours in vivo. In the present work, we try to summarize the data showing that some elements of the Phosphoinositide 3-kinase Class I (PI3K)/ Thymoma viral oncogene protein kinase (Akt) pathway, such the activity of PI3K Class I or Akt2, are necessary to maintain the CSC-like phenotype as well as survival of CSCs (also denoted as tumour-initiating cells (TICs)). Our data and other laboratory data permit a working hypothesis in which each Akt isoform plays an important and specific role in CSC/TIC growth, self-renewal, maintaining survival, and epithelial-mesenchymal transition (EMT) phenotype, not only in breast cancer, but also in glioma. We suggest that a more complete understanding is needed of the possible roles of isoforms in human tumours (iso-signalling determination). Thus, a comprehensive analysis of how hierarchical signalling is assembled during oncogenesis, how cancer landmarks are interconnected to favour CSC and tumour growth, and how some protein isoforms play a specific role in CSCs to ensure that survival and proliferation must be done in order to propose/generate new therapeutic approaches (alone or in combination with existing ones) to use against cancer.
Collapse
Affiliation(s)
- Sergio Rivas
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), 28049 Madrid, Spain.
- Centro Nacional de Biotecnología (CNB-CSIC), 28049 Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain.
| | - Carla Gómez-Oro
- Centro Nacional de Biotecnología (CNB-CSIC), 28049 Madrid, Spain.
| | - Inés M Antón
- Centro Nacional de Biotecnología (CNB-CSIC), 28049 Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain.
| | - Francisco Wandosell
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), 28049 Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain.
| |
Collapse
|
65
|
Renal Tubule Repair: Is Wnt/β-Catenin a Friend or Foe? Genes (Basel) 2018; 9:genes9020058. [PMID: 29364168 PMCID: PMC5852554 DOI: 10.3390/genes9020058] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/14/2018] [Accepted: 01/16/2018] [Indexed: 12/15/2022] Open
Abstract
Wnt/β-catenin signaling is extremely important for proper kidney development. This pathway is also upregulated in injured renal tubular epithelia, both in acute kidney injury and chronic kidney disease. The renal tubular epithelium is an important target of kidney injury, and its response (repair versus persistent injury) is critical for determining whether tubulointerstitial fibrosis, the hallmark of chronic kidney disease, develops. This review discusses how Wnt/β-catenin signaling in the injured tubular epithelia promotes either repair or fibrosis after kidney injury. There is data suggesting that epithelial Wnt/β-catenin signaling is beneficial in acute kidney injury and important in tubular progenitors responsible for epithelial repair. The role of Wnt/β-catenin signaling in chronically injured epithelia is less clear. There is convincing data that Wnt/β-catenin signaling in interstitial fibroblasts and pericytes contributes to the extracellular matrix accumulation that defines fibrosis. However, some recent studies question whether Wnt/β-catenin signaling in chronically injured epithelia actually promotes fibrosis or repair.
Collapse
|
66
|
van Doeselaar S, Burgering BMT. FOXOs Maintaining the Equilibrium for Better or for Worse. Curr Top Dev Biol 2018; 127:49-103. [PMID: 29433740 DOI: 10.1016/bs.ctdb.2017.10.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
A paradigm shift is emerging within the FOXO field and accumulating evidence indicates that we need to reappreciate the role of FOXOs, at least in cancer development. Here, we discuss the possibility that FOXOs are both tumor suppressors as well as promoters of tumor progression. This is mostly dependent on the biological context. Critical to this dichotomous role is the notion that FOXOs are central in preserving cellular homeostasis in redox control, genomic stability, and protein turnover. From this perspective, a paradoxical role in both suppressing and enhancing tumor progression can be reconciled. As many small molecules targeting the PI3K pathway are developed by big pharmaceutical companies and/or are in clinical trial, we will discuss what the consequences may be for the context-dependent role of FOXOs in tumor development in treatment options based on active PI3K signaling in tumors.
Collapse
Affiliation(s)
- Sabina van Doeselaar
- Molecular Cancer Research, Center Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Boudewijn M T Burgering
- Molecular Cancer Research, Center Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
67
|
Zhu L, Baker SS, Shahein A, Choudhury S, Liu W, Bhatia T, Baker RD, Lee T. Upregulation of non-canonical Wnt ligands and oxidative glucose metabolism in NASH induced by methionine-choline deficient diet. TRENDS IN CELL & MOLECULAR BIOLOGY 2018; 13:47-56. [PMID: 30853754 PMCID: PMC6407712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Wnt ligands regulate metabolic pathways, and dysregulation of Wnt signaling contributes to chronic inflammatory disease. A knowledge gap exists concerning the role of aberrant Wnt signaling in non-alcoholic steatohepatitis (NASH), which exhibits metabolic syndrome and inflammation. Using a mouse model of methionine-choline deficient diet (MCDD)-induced NASH, we investigated the Wnt signaling pathways in relation to hepatic glucose oxidation. Mice fed the MCD diet for 6 weeks developed prominent NASH marked by macrovesicular steatosis, inflammation and lipid peroxidation. qPCR analysis reveals differential hepatic expression of canonical and non-canonical Wnt ligands. While expression of Wnt3a was decreased in NASH vs chow diet control, expression of Wnt5a and Wnt11 were increased 3 fold and 15 fold, respectively. Consistent with activation of non-canonical Wnt signaling, expression of the alternative Wnt receptor ROR2 was increased 5 fold with no change in LRP6 expression. Activities of the metabolic enzymes glucokinase, phosphoglucoisomerase, glyceraldehyde-3-phosphate dehydrogenase, pyruvate kinase, and pyruvate dehydrogenase were all elevated by MCDD. NASH-driven glucose oxidation was accompanied by a 6-fold increase in lactate dehydrogenase (LDH)-B with no change in LDH-A. In addition, glucose-6-phosphate dehydrogenase, the regulatory and NADPH-producing enzyme of the pentose phosphate pathway, was elevated in NASH. These data support a role of accelerated glucose oxidation in the development of NASH, which may be driven by non-canonical Wnt signaling.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Techung Lee
- Corresponding author: Dr. Techung Lee, Tel: (716) 829-3106, , Department of Biochemistry, University at Buffalo, 955 Main Street, Buffalo, NY 14203
| |
Collapse
|
68
|
Russell JO, Monga SP. Wnt/β-Catenin Signaling in Liver Development, Homeostasis, and Pathobiology. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 13:351-378. [PMID: 29125798 DOI: 10.1146/annurev-pathol-020117-044010] [Citation(s) in RCA: 291] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The liver is an organ that performs a multitude of functions, and its health is pertinent and indispensable to survival. Thus, the cellular and molecular machinery driving hepatic functions is of utmost relevance. The Wnt signaling pathway is one such signaling cascade that enables hepatic homeostasis and contributes to unique hepatic attributes such as metabolic zonation and regeneration. The Wnt/β-catenin pathway plays a role in almost every facet of liver biology. Furthermore, its aberrant activation is also a hallmark of various hepatic pathologies. In addition to its signaling function, β-catenin also plays a role at adherens junctions. Wnt/β-catenin signaling also influences the function of many different cell types. Due to this myriad of functions, Wnt/β-catenin signaling is complex, context-dependent, and highly regulated. In this review, we discuss the Wnt/β-catenin signaling pathway, its role in cell-cell adhesion and liver function, and the cell type-specific roles of Wnt/β-catenin signaling as it relates to liver physiology and pathobiology.
Collapse
Affiliation(s)
- Jacquelyn O Russell
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | - Satdarshan P Monga
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania 15261, USA.,Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania 15261, USA.,Pittsburgh Liver Research Center, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania 15261, USA;
| |
Collapse
|
69
|
Abstract
Obesity has been estimated to decrease life expectancy by as little as 0.8 to as much as 7 years being the second leading cause of preventable death in the United States after smoking. Along with the increase in the prevalence of obesity, there has been a dramatic rise of the prevalence of prediabetes and type 2 diabetes among adolescents. Despite that, very little is known about the pathogenesis of these conditions in pediatrics and about how we could detect prediabetes in an early stage in order to prevent full blown diabetes. In this review we summarize the current knowledge on the pathophysiology of prediabetes and type 2 diabetes in adolescents and describe how biomarkers of beta-cell function might help identifying those individuals who are prone to progress from normal glucose tolerance towards prediabetes and overt type 2 diabetes. To better understand and fight this disease, we will need to explore and develop novel therapeutic strategies and individuate more sensitive and specific biomarkers that can allow an earlier detection of the disease.
Collapse
|
70
|
Cui ZH, Yuan Q, Mao L, Chen FL, Ji F, Tao S. Insulin resistance in vitamin D-deficient mice is alleviated by n-acetylcysteine. Oncotarget 2017; 8:63281-63289. [PMID: 28968988 PMCID: PMC5609920 DOI: 10.18632/oncotarget.18793] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 05/23/2017] [Indexed: 01/11/2023] Open
Abstract
Vitamin D deficiency will lead to insulin resistance. In the current study, vitamin D3 1α-Hydroxylase [“1α(OH)ase”] knockout mice were generated to mimic vitamin D deficiency in vivo. As compared to the wild-type mice, the liver tissues of the knockout mice showed impaired insulin signaling, decreased glucose transporter 4 expression and increased reactive oxygen species production. Meanwhile, p53-p21 activation, apoptosis intensity and pro-inflammatory cytokines (IL-6, IL-1 and MIP-1α) level were significantly increased in the knockout mice livers. Significantly, such effects in the knockout mice were largely attenuated by supplement with anti-oxidant n-acetylcysteine (NAC). Remarkably, insulin resistance and metabolic abnormalities in the knockout mice were largely alleviated after treatment of NAC. Therefore, inhibition of oxidative stress by NAC alleviated insulin resistance in vitamin D-deficient mice. Oxidative stress could be the primary cause of insulin resistance by vitamin D deficiency.
Collapse
Affiliation(s)
- Zhao-Hui Cui
- Department of Endocrinology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, China
| | - Qi Yuan
- Department of Endocrinology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, China
| | - Li Mao
- Department of Endocrinology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, China
| | - Feng-Li Chen
- Clinical Laboratory, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, China
| | - Feng Ji
- Department of Orthopedics, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, China
| | - Sha Tao
- Department of Endocrinology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, China
| |
Collapse
|
71
|
Soto-Gutierrez A, Gough A, Vernetti LA, Taylor DL, Monga SP. Pre-clinical and clinical investigations of metabolic zonation in liver diseases: The potential of microphysiology systems. Exp Biol Med (Maywood) 2017; 242:1605-1616. [PMID: 28467181 DOI: 10.1177/1535370217707731] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The establishment of metabolic zonation within a hepatic lobule ascribes specific functions to hepatocytes based on unique, location-dependent gene expression patterns. Recently, there have been significant developments in the field of metabolic liver zonation. A little over a decade ago, the role of β-catenin signaling was identified as a key regulator of gene expression and function in pericentral hepatocytes. Since then, additional molecules have been identified that regulate the pattern of Wnt/β-catenin signaling within a lobule and determine gene expression and function in other hepatic zones. Currently, the molecular basis of metabolic zonation in the liver appears to be a 'push and pull' between signaling pathways. Such compartmentalization not only provides an efficient assembly line for hepatocyte functions but also can account for restricting the initial hepatic damage and pathology from some hepatotoxic drugs to specific zones, possibly enabling effective regeneration and restitution responses from unaffected cells. Careful analysis and experimentation have also revealed that many pathological conditions in the liver lobule are spatially heterogeneous. We will review current research efforts that have focused on examination of the role and regulation of such mechanisms of hepatocyte adaptation and repair. We will discuss how the pathological organ-specific microenvironment affects cell signaling and metabolic liver zonation, especially in steatosis, viral hepatitis, and hepatocellular carcinoma. We will discuss how the use of new human microphysiological platforms will lead to a better understanding of liver disease progression, diagnosis, and therapies. In conclusion, we aim to provide insights into the role and regulation of metabolic zonation and function using traditional and innovative approaches. Impact statement Liver zonation of oxygen tension along the liver sinusoids has been identified as a critical liver microenvironment that impacts specific liver functions such as intermediary metabolism of amino acids, lipids, and carbohydrates, detoxification of xenobiotics and as sites for initiation of liver diseases. To date, most information on the role of zonation in liver disease including, non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), cirrhosis, and hepatocellular carcinoma (HCC) have been obtained from animal models. It is now possible to complement animal studies with human liver, microphysiology systems (MPS) containing induced pluripotent stem cells engineered to create disease models where it is also possible to control the in vitro liver oxygen microenvironment to define the role of zonation on the mechanism(s) of disease progression. The field now has the tools to investigate human liver disease progression, diagnosis, and therapeutic development.
Collapse
Affiliation(s)
| | - Albert Gough
- 2 Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA.,3 Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Lawrence A Vernetti
- 2 Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA.,3 Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - D L Taylor
- 2 Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA.,3 Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA.,4 Cancer Institute, University of Pittsburgh, Pittsburgh PA 15232, USA
| | - Satdarshan P Monga
- 1 Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15260, USA.,5 Department of Medicine, Pittsburgh, University of Pittsburgh, PA 15260, USA
| |
Collapse
|
72
|
Bonnet N. Bone-Derived Factors: A New Gateway to Regulate Glycemia. Calcif Tissue Int 2017; 100:174-183. [PMID: 27832316 DOI: 10.1007/s00223-016-0210-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 11/02/2016] [Indexed: 01/26/2023]
Abstract
Type 2 diabetes mellitus (T2DM) and osteoporosis are two major disorders which prevalence increases with aging and is predicted to worsen in the coming years. Preclinical investigations suggest common mechanisms implicated in the pathogenesis of both disorders. Recent evidence has established that there is a clear link between glucose and bone metabolism. The emergence of bone as an endocrine regulator through FGF23 and osteocalcin has led to the re-evaluation of the role of bone cells and bone-derived factors in the development of metabolic diseases such as T2DM. The development of bone morphogenetic proteins, fibroblast growth factor 23, and osteoprotegerin-deficient mice has allowed to elucidate their role in bone homeostasis, as well as revealed their potential important function in glucose homeostasis. This review proposes emerging perspectives for several bone-derived factors that may regulate glycemia through the activation or inhibition of bone remodeling or directly by regulating function of key organs such as pancreatic beta cell proliferation, insulin expression and secretion, storage and release of glucose from the liver, skeletal muscle contraction, and browning of the adipose tissue. Connections between organs including bone-derived factors should further be explored to understand the pathophysiology of glucose metabolism and diabetes.
Collapse
Affiliation(s)
- Nicolas Bonnet
- Division of Bone Diseases, Department of Internal Medicine Specialties, Geneva University Hospitals and Faculty of Medicine, 64 Av de la Roseraie, 1205, Geneva 14, Switzerland.
| |
Collapse
|
73
|
De Ita-Pérez DL, Díaz-Muñoz M. Synchronization by Daytime Restricted Food Access Modulates the Presence and Subcellular Distribution of β-Catenin and Its Phosphorylated Forms in the Rat Liver. Front Endocrinol (Lausanne) 2017; 8:14. [PMID: 28220106 PMCID: PMC5292920 DOI: 10.3389/fendo.2017.00014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 01/16/2017] [Indexed: 12/31/2022] Open
Abstract
β-catenin, the principal effector of the Wnt pathway, is also one of the cadherin cell adhesion molecules; therefore, it fulfills signaling and structural roles in most of the tissues and organs. It has been reported that β-catenin in the liver regulates metabolic responses such as gluconeogenesis and histological changes in response to obesity-promoting diets. The function and cellular location of β-catenin is finely modulated by coordinated sequences of phosphorylation-dephosphorylation events. In this article, we evaluated the levels and cellular localization of liver β-catenin variants, more specifically β-catenin phosphorylated in serine 33 (this phosphorylation provides recognizing sites for β-TrCP, which results in ubiquitination and posterior proteasomal degradation of β-catenin) and β-catenin phosphorylated in serine 675 (phosphorylation that enhances signaling and transcriptional activity of β-catenin through recruitment of different transcriptional coactivators). β-catenin phosphorylated in serine 33 in the nucleus shows day-night fluctuations in their expression level in the Ad Libitum group. In addition, we used a daytime restricted feeding (DRF) protocol to show that the above effects are sensitive to food access-dependent circadian synchronization. We found through western blot and immunohistochemical analyses that DRF protocol promoted (1) higher total β-catenins levels mainly associated with the plasma membrane, (2) reduced the presence of cytoplasmic β-catenin phosphorylated in serine 33, (3) an increase in nuclear β-catenin phosphorylated in serine 675, (4) differential co-localization of total β-catenins/β-catenin phosphorylated in serine 33 and total β-catenins/β-catenin phosphorylated in serine 675 at different temporal points along day and in fasting and refeeding conditions, and (5) differential liver zonation of β-catenin variants studied along hepatic acinus. In conclusion, the present data comprehensively characterize the effect food synchronization has on the presence, subcellular distribution, and liver zonation of β-catenin variants. These results are relevant to understand the set of metabolic and structural liver adaptations that are associated with the expression of the food entrained oscillator (FEO).
Collapse
Affiliation(s)
- Dalia Luz De Ita-Pérez
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Querétaro, México
| | - Mauricio Díaz-Muñoz
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Querétaro, México
- *Correspondence: Mauricio Díaz-Muñoz,
| |
Collapse
|
74
|
Altered DNA methylation associated with an abnormal liver phenotype in a cattle model with a high incidence of perinatal pathologies. Sci Rep 2016; 6:38869. [PMID: 27958319 PMCID: PMC5153653 DOI: 10.1038/srep38869] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 11/14/2016] [Indexed: 02/06/2023] Open
Abstract
Cloning enables the generation of both clinically normal and pathological individuals from the same donor cells, and may therefore be a DNA sequence-independent driver of phenotypic variability. We took advantage of cattle clones with identical genotypes but different developmental abilities to investigate the role of epigenetic factors in perinatal mortality, a complex trait with increasing prevalence in dairy cattle. We studied livers from pathological clones dying during the perinatal period, clinically normal adult clones with the same genotypes as perinatal clones and conventional age-matched controls. The livers from deceased perinatal clones displayed histological lesions, modifications to quantitative histomorphometric and metabolic parameters such as glycogen storage and fatty acid composition, and an absence of birth-induced maturation. In a genome-wide epigenetic analysis, we identified DNA methylation patterns underlying these phenotypic alterations and targeting genes relevant to liver metabolism, including the type 2 diabetes gene TCF7L2. The adult clones were devoid of major phenotypic and epigenetic abnormalities in the liver, ruling out the effects of genotype on the phenotype observed. These results thus provide the first demonstration of a genome-wide association between DNA methylation and perinatal mortality in cattle, and highlight epigenetics as a driving force for phenotypic variability in farmed animals.
Collapse
|
75
|
Kulebyakin K, Penkov D, Blasi F, Akopyan Z, Tkachuk V. The transcription factor Prep1 controls hepatic insulin sensitivity and gluconeogenesis by targeting nuclear localization of FOXO1. Biochem Biophys Res Commun 2016; 481:182-188. [PMID: 27815072 DOI: 10.1016/j.bbrc.2016.10.146] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 10/29/2016] [Indexed: 10/20/2022]
Abstract
Liver plays a key role in controlling body carbohydrate homeostasis by switching between accumulation and production of glucose and this way maintaining constant level of glucose in blood. Increased blood glucose level triggers release of insulin from pancreatic β-cells. Insulin represses hepatic glucose production and increases glucose accumulation. Insulin resistance is the main cause of type 2 diabetes and hyperglycemia. Currently thiazolidinediones (TZDs) targeting transcriptional factor PPARγ are used as insulin sensitizers for treating patients with type 2 diabetes. However, TZDs are reported to be associated with cardiovascular and liver problems and stimulate obesity. Thus, it is necessary to search new approaches to improve insulin sensitivity. A promising candidate is transcriptional factor Prep1, as it was shown earlier it could affect insulin sensitivity in variety of insulin-sensitive tissues. The aim of the present study was to evaluate a possible involvement of transcriptional factor Prep1 in control of hepatic glucose accumulation and production. We created mice with liver-specific Prep1 knockout and discovered that hepatocytes derived from these mice are much more sensitive to insulin, comparing to their WT littermates. Incubation of these cells with 100 nM insulin results in almost complete inhibition of gluconeogenesis, while in WT cells this repression is only partial. However, Prep1 doesn't affect gluconeogenesis in the absence of insulin. Also, we observed that nuclear content of gluconeogenic transcription factor FOXO1 was greatly reduced in Prep1 knockout hepatocytes. These findings suggest that Prep1 may control hepatic insulin sensitivity by targeting FOXO1 nuclear stability.
Collapse
Affiliation(s)
- Konstantin Kulebyakin
- Lomonosov Moscow State University, Faculty of Fundamental Medicine, Department of Biochemistry and Molecular Medicine, Lomonosovsky Prospekt 31-5, Moscow, 117192, Russia.
| | - Dmitry Penkov
- Lomonosov Moscow State University, Faculty of Fundamental Medicine, Department of Biochemistry and Molecular Medicine, Lomonosovsky Prospekt 31-5, Moscow, 117192, Russia; IFOM - the FIRC Institute of Molecular Oncology, Via Adamello 16, Milan, 20139, Italy
| | - Francesco Blasi
- IFOM - the FIRC Institute of Molecular Oncology, Via Adamello 16, Milan, 20139, Italy
| | - Zhanna Akopyan
- Lomonosov Moscow State University, Faculty of Fundamental Medicine, Department of Biochemistry and Molecular Medicine, Lomonosovsky Prospekt 31-5, Moscow, 117192, Russia
| | - Vsevolod Tkachuk
- Lomonosov Moscow State University, Faculty of Fundamental Medicine, Department of Biochemistry and Molecular Medicine, Lomonosovsky Prospekt 31-5, Moscow, 117192, Russia
| |
Collapse
|
76
|
Zhang Q, Sun X, Xiao X, Zheng J, Li M, Yu M, Ping F, Wang Z, Qi C, Wang T, Wang X. Maternal Chromium Restriction Leads to Glucose Metabolism Imbalance in Mice Offspring through Insulin Signaling and Wnt Signaling Pathways. Int J Mol Sci 2016; 17:ijms17101767. [PMID: 27782077 PMCID: PMC5085791 DOI: 10.3390/ijms17101767] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 10/08/2016] [Accepted: 10/17/2016] [Indexed: 12/13/2022] Open
Abstract
An adverse intrauterine environment, induced by a chromium-restricted diet, is a potential cause of metabolic disease in adult life. Up to now, the relative mechanism has not been clear. C57BL female mice were time-mated and fed either a control diet (CD), or a chromium-restricted diet (CR) throughout pregnancy and the lactation period. After weaning, some offspring continued the diet diagram (CD-CD or CR-CR), while other offspring were transferred to another diet diagram (CD-CR or CR-CD). At 32 weeks of age, glucose metabolism parameters were measured, and the liver from CR-CD group and CD-CD group was analyzed using a gene array. Quantitative real-time polymerase chain reaction (qPCR) and Western blot were used to verify the result of the gene array. A maternal chromium-restricted diet resulted in obesity, hyperglycemia, hyperinsulinemia, increased area under the curve (AUC) of glucose in oral glucose tolerance testing and homeostasis model assessment of insulin resistance (HOMA-IR). There were 463 genes that differed significantly (>1.5-fold change, p < 0.05) between CR-CD offspring (264 up-regulated genes, 199 down-regulated genes) and control offspring. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and STRING (Search Tool for the Retrieval of Interacting Genes/Proteins) analysis revealed that the insulin signaling pathway and Wnt signaling pathway were in the center of the gene network. Our study provides the first evidence that maternal chromium deficiency influences glucose metabolism in pups through the regulation of insulin signaling and Wnt signaling pathways.
Collapse
Affiliation(s)
- Qian Zhang
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Xiaofang Sun
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| | - Xinhua Xiao
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Jia Zheng
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Ming Li
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Miao Yu
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Fan Ping
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Zhixin Wang
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Cuijuan Qi
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Tong Wang
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Xiaojing Wang
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|
77
|
Christensen LL, True K, Hamilton MP, Nielsen MM, Damas ND, Damgaard CK, Ongen H, Dermitzakis E, Bramsen JB, Pedersen JS, Lund AH, Vang S, Stribolt K, Madsen MR, Laurberg S, McGuire SE, Ørntoft TF, Andersen CL. SNHG16 is regulated by the Wnt pathway in colorectal cancer and affects genes involved in lipid metabolism. Mol Oncol 2016; 10:1266-82. [PMID: 27396952 PMCID: PMC5423192 DOI: 10.1016/j.molonc.2016.06.003] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 05/02/2016] [Accepted: 06/17/2016] [Indexed: 02/07/2023] Open
Abstract
It is well established that lncRNAs are aberrantly expressed in cancer where they have been shown to act as oncogenes or tumor suppressors. RNA profiling of 314 colorectal adenomas/adenocarcinomas and 292 adjacent normal colon mucosa samples using RNA-sequencing demonstrated that the snoRNA host gene 16 (SNHG16) is significantly up-regulated in adenomas and all stages of CRC. SNHG16 expression was positively correlated to the expression of Wnt-regulated transcription factors, including ASCL2, ETS2, and c-Myc. In vitro abrogation of Wnt signaling in CRC cells reduced the expression of SNHG16 indicating that SNHG16 is regulated by the Wnt pathway. Silencing of SNHG16 resulted in reduced viability, increased apoptotic cell death and impaired cell migration. The SNHG16 silencing particularly affected expression of genes involved in lipid metabolism. A connection between SNHG16 and genes involved in lipid metabolism was also observed in clinical tumors. Argonaute CrossLinking and ImmunoPrecipitation (AGO-CLIP) demonstrated that SNHG16 heavily binds AGO and has 27 AGO/miRNA target sites along its length, indicating that SNHG16 may act as a competing endogenous RNA (ceRNA) "sponging" miRNAs off their cognate targets. Most interestingly, half of the miRNA families with high confidence targets on SNHG16 also target the 3'UTR of Stearoyl-CoA Desaturase (SCD). SCD is involved in lipid metabolism and is down-regulated upon SNHG16 silencing. In conclusion, up-regulation of SNHG16 is a frequent event in CRC, likely caused by deregulated Wnt signaling. In vitro analyses demonstrate that SNHG16 may play an oncogenic role in CRC and that it affects genes involved in lipid metabolism, possible through ceRNA related mechanisms.
Collapse
Affiliation(s)
- Lise Lotte Christensen
- Department of Molecular Medicine (MOMA), Aarhus University Hospital, University of Aarhus, Aarhus, Denmark.
| | - Kirsten True
- Department of Molecular Medicine (MOMA), Aarhus University Hospital, University of Aarhus, Aarhus, Denmark.
| | - Mark P Hamilton
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| | - Morten M Nielsen
- Department of Molecular Medicine (MOMA), Aarhus University Hospital, University of Aarhus, Aarhus, Denmark.
| | - Nkerorema D Damas
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark.
| | | | - Halit Ongen
- Department of Genetic Medicine and Development, Functional Population Genomics and Genetics of Complex Traits Lab, University of Geneva Medical School, Geneva, Switzerland.
| | - Emmanouil Dermitzakis
- Department of Genetic Medicine and Development, Functional Population Genomics and Genetics of Complex Traits Lab, University of Geneva Medical School, Geneva, Switzerland.
| | - Jesper B Bramsen
- Department of Molecular Medicine (MOMA), Aarhus University Hospital, University of Aarhus, Aarhus, Denmark.
| | - Jakob S Pedersen
- Department of Molecular Medicine (MOMA), Aarhus University Hospital, University of Aarhus, Aarhus, Denmark.
| | - Anders H Lund
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark.
| | - Søren Vang
- Department of Molecular Medicine (MOMA), Aarhus University Hospital, University of Aarhus, Aarhus, Denmark.
| | - Katrine Stribolt
- Department of Pathology, Aarhus University Hospital, University of Aarhus, Aarhus, Denmark.
| | - Mogens R Madsen
- Surgical Research Unit, Herning Regional Hospital, Herning, Denmark.
| | - Søren Laurberg
- Department of Surgery, Aarhus University Hospital, University of Aarhus, Aarhus, Denmark.
| | - Sean E McGuire
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Torben F Ørntoft
- Department of Molecular Medicine (MOMA), Aarhus University Hospital, University of Aarhus, Aarhus, Denmark.
| | - Claus L Andersen
- Department of Molecular Medicine (MOMA), Aarhus University Hospital, University of Aarhus, Aarhus, Denmark.
| |
Collapse
|
78
|
Lin H, Angeli M, Chung KJ, Ejimadu C, Rosa AR, Lee T. sFRP2 activates Wnt/β-catenin signaling in cardiac fibroblasts: differential roles in cell growth, energy metabolism, and extracellular matrix remodeling. Am J Physiol Cell Physiol 2016; 311:C710-C719. [PMID: 27605451 DOI: 10.1152/ajpcell.00137.2016] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 08/29/2016] [Indexed: 02/06/2023]
Abstract
Secreted Frizzled-related protein 2 (sFRP2) plays a key role in chronic fibrosis after myocardial infarction and in heart failure. The aim of this study was to elucidate the mechanisms through which sFRP2 may regulate the growth and extracellular matrix (ECM) remodeling of adult mouse cardiac fibroblasts (CFs). We found that sFRP2 activates CFs in part through canonical Wnt/β-catenin signaling, as evidenced by increased expression of Axin2 and Wnt3a, but not Wnt5a, as well as accumulation of nuclear β-catenin. In response to sFRP2, CFs exhibited robust cell proliferation associated with increased glucose consumption and lactate production, a phenomenon termed "the Warburg effect" in oncology. The coupling between CF expansion and anaerobic glycolysis is marked by upregulation of glyceraldehyde-3-phosphate dehydrogenase and tissue-nonspecific alkaline phosphatase. In conjunction with these phenotypic changes, CFs accelerated ECM remodeling through upregulation of expression of the matrix metalloproteinase (MMP) 1 and MMP13 genes, two members of the collagenase subfamily, and enzyme activities of MMP2 and MMP9, two members of the gelatinase subfamily. Consistent with the induction of multiple MMPs possessing collagenolytic activities, the steady-state level of collagen type 1 in CF-spent medium was reduced by sFRP2. Analysis of non-CF cell types revealed that the multifaceted effects of sFRP2 on growth control, glucose metabolism, and ECM regulation are largely restricted to CFs and highly sensitive to Wnt signaling perturbation. The study provides a molecular framework on which the functional versatility and signaling complexity of sFRP2 in cardiac fibrosis may be better defined.
Collapse
Affiliation(s)
- Huey Lin
- Department of Biochemistry and Department of Biomedical Engineering, University at Buffalo, Buffalo, New York
| | - Mia Angeli
- Department of Biochemistry and Department of Biomedical Engineering, University at Buffalo, Buffalo, New York
| | - Kwang Jin Chung
- Department of Biochemistry and Department of Biomedical Engineering, University at Buffalo, Buffalo, New York
| | - Chukwuemeka Ejimadu
- Department of Biochemistry and Department of Biomedical Engineering, University at Buffalo, Buffalo, New York
| | - Angelica Rivera Rosa
- Department of Biochemistry and Department of Biomedical Engineering, University at Buffalo, Buffalo, New York
| | - Techung Lee
- Department of Biochemistry and Department of Biomedical Engineering, University at Buffalo, Buffalo, New York
| |
Collapse
|
79
|
Lin Y, Ohkawara B, Ito M, Misawa N, Miyamoto K, Takegami Y, Masuda A, Toyokuni S, Ohno K. Molecular hydrogen suppresses activated Wnt/β-catenin signaling. Sci Rep 2016; 6:31986. [PMID: 27558955 PMCID: PMC5001535 DOI: 10.1038/srep31986] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 08/01/2016] [Indexed: 01/23/2023] Open
Abstract
Molecular hydrogen (H2) is effective for many diseases. However, molecular bases of H2 have not been fully elucidated. Cumulative evidence indicates that H2 acts as a gaseous signal modulator. We found that H2 suppresses activated Wnt/β-catenin signaling by promoting phosphorylation and degradation οf β-catenin. Either complete inhibition of GSK3 or mutations at CK1- and GSK3-phosphorylation sites of β-catenin abolished the suppressive effect of H2. H2 did not increase GSK3-mediated phosphorylation of glycogen synthase, indicating that H2 has no direct effect on GSK3 itself. Knock-down of adenomatous polyposis coli (APC) or Axin1, which form the β-catenin degradation complex, minimized the suppressive effect of H2 on β-catenin accumulation. Accordingly, the effect of H2 requires CK1/GSK3-phosphorylation sites of β-catenin, as well as the β-catenin degradation complex comprised of CK1, GSK3, APC, and Axin1. We additionally found that H2 reduces the activation of Wnt/β-catenin signaling in human osteoarthritis chondrocytes. Oral intake of H2 water tended to ameliorate cartilage degradation in a surgery-induced rat osteoarthritis model through attenuating β-catenin accumulation. We first demonstrate that H2 suppresses abnormally activated Wnt/β-catenin signaling, which accounts for the protective roles of H2 in a fraction of diseases.
Collapse
Affiliation(s)
- Yingni Lin
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Bisei Ohkawara
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mikako Ito
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nobuaki Misawa
- Department of Pathology and Biological Responses, Graduate school of Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kentaro Miyamoto
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiko Takegami
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akio Masuda
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Graduate school of Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
80
|
Belinsky GS, Ward L, Chung C. Pigment epithelium-derived factor (PEDF) normalizes matrix defects in iPSCs derived from Osteogenesis imperfecta Type VI. Rare Dis 2016; 4:e1212150. [PMID: 27579219 PMCID: PMC4986704 DOI: 10.1080/21675511.2016.1212150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 06/14/2016] [Accepted: 07/06/2016] [Indexed: 01/28/2023] Open
Abstract
Osteogenesis imperfecta (OI) Type VI is characterized by a defect in bone mineralization, which results in multiple fractures early in life. Null mutations in the PEDF gene, Serpinf1, are the cause of OI VI. Whether PEDF restoration in a murine model of OI Type VI could improve bone mass and function was previously unknown. In Belinsky et al, we provided evidence that PEDF delivery enhanced bone mass and improved parameters of bone function in vivo. Further, we demonstrated that PEDF temporally inhibits Wnt signaling to enhance osteoblast differentiation. Here, we demonstrate that generation of induced pluripotent stem cells (iPSCs) from a PEDF null patient provides additional evidence for PEDF's role in regulating extracellular matrix proteins secreted from osteoblasts. PEDF null iPSCs have marked abnormalities in secreted matrix proteins, capturing a key feature of human OI Type VI, which were normalized by exogenous PEDF. Lastly, we place our recent findings within the broader context of PEDF biology and the developmental signaling pathways that are implicated in its actions.
Collapse
Affiliation(s)
- Glenn S Belinsky
- Department of Medicine, Yale University School of Medicine , New Haven, CT, USA
| | - Leanne Ward
- Children's Hospital of Eastern Ontario , Ottawa, Canada
| | - Chuhan Chung
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA; VA CT Healthcare System, West Haven, CT, USA
| |
Collapse
|
81
|
Abstract
The recent recognition of the clinical association between type 2 diabetes (T2D) and several types of human cancer has been further highlighted by reports of antidiabetic drugs treating or promoting cancer. At the cellular level, a plethora of molecules operating within distinct signaling pathways suggests cross-talk between the multiple pathways at the interface of the diabetes–cancer link. Additionally, a growing body of emerging evidence implicates homeostatic pathways that may become imbalanced during the pathogenesis of T2D or cancer or that become chronically deregulated by prolonged drug administration, leading to the development of cancer in diabetes and vice versa. This notion underscores the importance of combining clinical and basic mechanistic studies not only to unravel mechanisms of disease development but also to understand mechanisms of drug action. In turn, this may help the development of personalized strategies in which drug doses and administration durations are tailored to individual cases at different stages of the disease progression to achieve more efficacious treatments that undermine the diabetes–cancer association.
Collapse
Affiliation(s)
- Slavica Tudzarova
- Wolfson Institute for Biomedical Research, University College London, London WC1E6BT, UK
| | - Mahasin A Osman
- Department of Molecular Physiology, Pharmacology and Biotechnology, Division of Biology and Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912 Department of Chemistry and Forensic Sciences, College of Sciences and Technology, Savannah State University, Savannah, GA 41404
| |
Collapse
|
82
|
Zheng H, Miyakawa T, Sawano Y, Asano A, Okumura A, Yamagoe S, Tanokura M. Crystal Structure of Human Leukocyte Cell-derived Chemotaxin 2 (LECT2) Reveals a Mechanistic Basis of Functional Evolution in a Mammalian Protein with an M23 Metalloendopeptidase Fold. J Biol Chem 2016; 291:17133-42. [PMID: 27334921 DOI: 10.1074/jbc.m116.720375] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Indexed: 01/19/2023] Open
Abstract
Human leukocyte cell-derived chemotaxin 2 (LECT2), which is predominantly expressed in the liver, is a multifunctional protein. LECT2 is becoming a potential therapeutic target for several diseases of worldwide concern such as rheumatoid arthritis, hepatocellular carcinoma, and obesity. Here, we present the crystal structure of LECT2, the first mammalian protein whose structure contains an M23 metalloendopeptidase fold. The LECT2 structure adopts a conserved Zn(II) coordination configuration but lacks a proposed catalytic histidine residue, and its potential substrate-binding groove is blocked in the vicinity of the Zn(II)-binding site by an additional intrachain loop at the N terminus. Consistent with these structural features, LECT2 was found to be catalytically inactive as a metalloendopeptidase against various types of peptide sequences, including pentaglycine. In addition, a surface plasmon resonance analysis demonstrated that LECT2 bound to the c-Met receptor with micromolar affinity. These results indicate that LECT2 likely plays its critical roles by acting as a ligand for the corresponding protein receptors rather than as an enzymatically active peptidase. The intrachain loop together with the pseudo-active site groove in LECT2 structure may be specific for interactions between LECT2 and receptors. Our study reveals a mechanistic basis for the functional evolution of a mammalian protein with an M23 metalloendopeptidase fold and potentially broadens the implications for the biological importance of noncatalytic peptidases in the M23 family.
Collapse
Affiliation(s)
- Hai Zheng
- From the Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Takuya Miyakawa
- From the Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Yoriko Sawano
- From the Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan, Department of Chemistry, College of Liberal Arts and Sciences, Tokyo Medical and Dental University, 2-8-30 Kounodai, Ichikawa-shi, Chiba 272-0827, Japan
| | - Atsuko Asano
- From the Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Akinori Okumura
- Department of Diabetic Complications, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan, and
| | - Satoshi Yamagoe
- Department of Chemotherapy and Mycoses, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Masaru Tanokura
- From the Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan,
| |
Collapse
|
83
|
Jin T. Current Understanding on Role of the Wnt Signaling Pathway Effector TCF7L2 in Glucose Homeostasis. Endocr Rev 2016; 37:254-77. [PMID: 27159876 DOI: 10.1210/er.2015-1146] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The role of the Wnt signaling pathway in metabolic homeostasis has drawn our intensive attention, especially after the genome-wide association study discovery that certain polymorphisms of its key effector TCF7L2 are strongly associated with the susceptibility to type 2 diabetes. For a decade, great efforts have been made in determining the function of TCF7L2 in various metabolic organs, which have generated both considerable achievements and disputes. In this review, I will briefly introduce the canonical Wnt signaling pathway, focusing on its effector β-catenin/TCF, including emphasizing the bidirectional feature of TCFs and β-catenin post-translational modifications. I will then summarize the observations on the association between TCF7L2 polymorphisms and type 2 diabetes risk. The main content, however, is on the intensive functional exploration of the metabolic role of TCF7L2, including the disputes generated on determining its role in the pancreas and liver with various transgenic mouse lines. Finally, I will discuss those achievements and disputes and present my future perspectives.
Collapse
Affiliation(s)
- Tianru Jin
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada
| |
Collapse
|
84
|
Liu Z, Xiao Y, Zhou Z, Mao X, Cai J, Xiong L, Liao C, Huang F, Liu Z, Ali Sheikh MS, Plutzky J, Huang H, Yang T, Duan Q. Extensive metabolic disorders are present in APC(min) tumorigenesis mice. Mol Cell Endocrinol 2016; 427:57-64. [PMID: 26948948 DOI: 10.1016/j.mce.2016.03.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Revised: 02/29/2016] [Accepted: 03/03/2016] [Indexed: 01/08/2023]
Abstract
Wnt signaling plays essential role in mesenchymal stem cell (MSC) differentiation. Activation of Wnt signaling suppresses adipogenesis, but promotes osteogenesis in MSC. Adenomatous polyposis coli (APC) is a negative regulator of β-catenin and Wnt signaling activity. The mutation of APC gene leads to the activation of Wnt signaling and is responsible for tumorigenesis in APC(min) mouse; however, very few studies focused on its metabolic abnormalities. The present study reports a widespread metabolic disorder phenotype in APC(min) mice. The old APC(min) mice have decreased body weight and impaired adipogenesis, but severe hyperlipidemia, which mimic the phenotypes of Familial Adenomatous Polyposis (FAP), an inherited disease also caused by APC gene mutation in human. We found that the expression of lipid metabolism and free fat acids (FA) use genes in the white adipose tissue (WAT) of the APC(min) mice is much lower than those of control. The changed gene expression pattern may lead to the disability of circulatory lipid transportation and storage at WAT. Moreover, the APC(min) mice could not maintain the core body temperature in cold condition. PET-CT determination revealed that the BAT of APC(min) mice has significantly impaired ability to take up (18)FDG from the blood. Morphological studies identified that the brown adipocytes of APC(min) mice were filled with lipid droplets but fewer mitochondria. These results matched with the findings of impaired BAT function in APC(min) mice. Collectively, our study explores a new mechanism that explains abnormal metabolism in APC(min) mice and provides insights into studying the metabolic disorders of FAP patients.
Collapse
Affiliation(s)
- Zhenzhen Liu
- Cardiovascular Division, Xiangya Hospital, Central South University, Changsha, China
| | - Yi Xiao
- Cardiovascular Division, Xiangya Hospital, Central South University, Changsha, China
| | - Zhengxiang Zhou
- Cardiovascular Division, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoxiao Mao
- Cardiovascular Division, Xiangya Hospital, Central South University, Changsha, China; Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jinxing Cai
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Lu Xiong
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Chaonan Liao
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Fulian Huang
- Department of Anatomy and Neuroscience, Xiangya School of Medicine, Central South University, Changsha, China
| | - Zehao Liu
- Endocrinology Division, Xiangya Hospital, Central South University, Changsha, China
| | - Md Sayed Ali Sheikh
- Cardiovascular Division, Xiangya Hospital, Central South University, Changsha, China
| | - Jorge Plutzky
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - He Huang
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, China.
| | - Tianlun Yang
- Cardiovascular Division, Xiangya Hospital, Central South University, Changsha, China.
| | - Qiong Duan
- Cardiovascular Division, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
85
|
Okabe H, Kinoshita H, Imai K, Nakagawa S, Higashi T, Arima K, Uchiyama H, Ikegami T, Harimoto N, Itoh S, Ishiko T, Yoshizumi T, Beppu T, Monga SPS, Baba H, Maehara Y. Diverse Basis of β-Catenin Activation in Human Hepatocellular Carcinoma: Implications in Biology and Prognosis. PLoS One 2016; 11:e0152695. [PMID: 27100093 PMCID: PMC4839611 DOI: 10.1371/journal.pone.0152695] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 03/17/2016] [Indexed: 01/20/2023] Open
Abstract
Aim β-catenin signaling is a major oncogenic pathway in hepatocellular carcinoma (HCC). Since β-catenin phosphorylation by glycogen synthase kinase 3β (GSK3β) and casein kinase 1ε (CK1ε) results in its degradation, mutations affecting these phosphorylation sites cause β-catenin stabilization. However, the relevance of missense mutations in non-phosphorylation sites in exon 3 remains unclear. The current study explores significance of such mutations in addition to addressing the clinical and biological implications of β-catenin activation in human HCC. Methods Gene alteration in exon3 of CTNNB1, gene expression of β-catenin targets such as glutamate synthetase (GS), axin2, lect2 and regucalcin (RGN), and protein expression of β-catenin were examined in 125 human HCC tissues. Results Sixteen patients (12.8%) showed conventional missense mutations affecting codons 33, 37, 41, and 45. Fifteen additional patients (12.0%) had other missense mutations in codon 32, 34, and 35. Induction of exon3 mutation caused described β-catenin target gene upregulation in HCC cell line. Interestingly, conventional and non-phosphorylation site mutations were equally associated with upregulation of β-catenin target genes. Nuclear localization of β-catenin was associated with poor overall survival (p = 0.0461). Of these patients with nuclear β-catenin localization, loss of described β-catenin target gene upregulation showed significant poorer overall survival than others (p = 0.0001). Conclusion This study suggests that both conventional and other missense mutations in exon 3 of CTNNB1 lead to β-catenin activation in human HCC. Additionally, the mechanism of nuclear β-catenin localization without upregulation of described β-catenin target genes might be of clinical importance depending on distinct mechanism.
Collapse
Affiliation(s)
- Hirohisa Okabe
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Hiroki Kinoshita
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Katsunori Imai
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Shigeki Nakagawa
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takaaki Higashi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kota Arima
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hideaki Uchiyama
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toru Ikegami
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Norifumi Harimoto
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinji Itoh
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takatoshi Ishiko
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Tomoharu Yoshizumi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toru Beppu
- Department of Multidisciplinary Treatment for Gastroenterological Cancer, Kumamoto University Hospital, Kumamoto, Japan
| | - Satdarshan P. S. Monga
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
- * E-mail:
| | - Yoshihiko Maehara
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
86
|
Personalized therapy for hepatocellular carcinoma: Where are we now? Cancer Treat Rev 2016; 45:77-86. [DOI: 10.1016/j.ctrv.2016.02.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Revised: 02/22/2016] [Accepted: 02/23/2016] [Indexed: 12/14/2022]
|
87
|
Ma Y, Lv X, He J, Liu T, Wen S, Wang L. Wnt agonist stimulates liver regeneration after small-for-size liver transplantation in rats. Hepatol Res 2016; 46:E154-64. [PMID: 26176339 DOI: 10.1111/hepr.12553] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 06/15/2015] [Accepted: 07/06/2015] [Indexed: 01/05/2023]
Abstract
AIM Liver regeneration is inhibited in small-for-size grafts, which plays a role in the failure of partial liver grafts after transplantation. The Wnt/β-catenin signaling pathway plays a critical role in liver development, regeneration and homeostasis. In this study, we investigated whether pharmacological activation of Wnt signaling improves liver regeneration after small-for-size liver transplantation. METHODS The livers of male Sprague-Dawley rats were reduced to approximately 50% and 30% of their original sizes and transplanted. A Wnt agonist (2-amino-4-[3,4-[methylenedioxy]benzylamino]-6-[3-methoxyphenyl] pyrimidine], 5 mg/kg bodyweight) or an equal volume of vehicle was administrated i.p. into the donor 1 h before the transplantation. Tissue and blood samples were collected at various times after transplantation, and a survival study was performed. RESULTS Hepatic expression of active β-catenin and its downstream target gene Axin2 were decreased in 30% of liver grafts after transplantation while the Wnt agonist increased their expression similar to the 50% liver grafts. The Wnt agonist reversed inhibition of cyclin D1 expression and adenosine triphosphate production in the 30% liver grafts compared with the 50% grafts. The Wnt agonist also attenuated hepatocellular injury and increased the hepatocyte proliferation response, liver regeneration rate and survival after transplantation of the 30% liver graft. CONCLUSION Activation of Wnt/β-catenin signaling in liver grafts by pharmacological pretreatment can accelerate regeneration in a partial liver transplant model.
Collapse
Affiliation(s)
- Yuefeng Ma
- Department of General Surgery, The Affiliated Zhongshan Hospital of Dalian University, Dalian, China.,Organ Transplantation Center, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiangwei Lv
- Organ Transplantation Center, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jinjing He
- Organ Transplantation Center, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Tianqing Liu
- Department of Pathology, Friendship Hospital of Dalian Medical University, Dalian, China
| | - Shuang Wen
- Department of Pathology, Friendship Hospital of Dalian Medical University, Dalian, China
| | - Liming Wang
- Organ Transplantation Center, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
88
|
Liu D, Mai K, Zhang Y, Xu W, Ai Q. Tumour necrosis factor-α inhibits hepatic lipid deposition through GSK-3β/β-catenin signaling in juvenile turbot (Scophthalmus maximus L.). Gen Comp Endocrinol 2016; 228:1-8. [PMID: 26747182 DOI: 10.1016/j.ygcen.2015.12.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 12/14/2015] [Accepted: 12/29/2015] [Indexed: 12/13/2022]
Abstract
In this study, the mechanism that TNFα inhibits lipid deposition through GSK-3β/β-catenin signaling was investigated in the liver of juvenile turbot (Scophthalmus maximus L.) by injection of TNFα or TNFα inhibitor pomalidomide (POM). It was found that TNFα inhibited the expression of GSK-3β and induced β-catenin expression. TNFα inhibited the expression of peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer binding protein α (C/EBPα), as well as the activity of lipoprotein lipase (LPL) and fatty acid synthetase (FAS). In addition, the level of triglyceride (TG), total cholesterol (TC), nonesterified fatty acid (NEFA), and glycerol was decreased by TNFα treatment in the liver. In the plasma, the level of TG, TC, low density lipoprotein cholesterol (LDL-C), NEFA, and glycerol was decreased, but high density lipoprotein cholesterol (HDL-C) was increased by TNFα treatment. However, compared to TNFα, POM had the opposite effect on the biochemical indexes and genes related to lipid deposition in the liver. The results indicated that TNFα may regulate hepatic lipid metabolism and fat distribution through GSK-3β/β-catenin signaling as well as transcription factors PPARγ and C/EBPα in juvenile turbot.
Collapse
Affiliation(s)
- Dongwu Liu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture) and the Key Laboratory of Mariculture (Education Ministry of China), Ocean University of China, Qingdao 266003, PR China
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture) and the Key Laboratory of Mariculture (Education Ministry of China), Ocean University of China, Qingdao 266003, PR China
| | - Yanjiao Zhang
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture) and the Key Laboratory of Mariculture (Education Ministry of China), Ocean University of China, Qingdao 266003, PR China
| | - Wei Xu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture) and the Key Laboratory of Mariculture (Education Ministry of China), Ocean University of China, Qingdao 266003, PR China
| | - Qinghui Ai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture) and the Key Laboratory of Mariculture (Education Ministry of China), Ocean University of China, Qingdao 266003, PR China.
| |
Collapse
|
89
|
Chang HR, Nam S, Kook MC, Kim KT, Liu X, Yao H, Jung HR, Lemos R, Seo HH, Park HS, Gim Y, Hong D, Huh I, Kim YW, Tan D, Liu CG, Powis G, Park T, Liang H, Kim YH. HNF4α is a therapeutic target that links AMPK to WNT signalling in early-stage gastric cancer. Gut 2016; 65:19-32. [PMID: 25410163 PMCID: PMC4717359 DOI: 10.1136/gutjnl-2014-307918] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 10/25/2014] [Indexed: 12/24/2022]
Abstract
BACKGROUND Worldwide, gastric cancer (GC) is the fourth most common malignancy and the most common cancer in East Asia. Development of targeted therapies for this disease has focused on a few known oncogenes but has had limited effects. OBJECTIVE To determine oncogenic mechanisms and novel therapeutic targets specific for GC by identifying commonly dysregulated genes from the tumours of both Asian-Pacific and Caucasian patients. METHODS We generated transcriptomic profiles of 22 Caucasian GC tumours and their matched non-cancerous samples and performed an integrative analysis across different GC gene expression datasets. We examined the inhibition of commonly overexpressed oncogenes and their constituent signalling pathways by RNAi and/or pharmacological inhibition. RESULTS Hepatocyte nuclear factor-4α (HNF4α) upregulation was a key signalling event in gastric tumours from both Caucasian and Asian patients, and HNF4α antagonism was antineoplastic. Perturbation experiments in GC tumour cell lines and xenograft models further demonstrated that HNF4α is downregulated by AMPKα signalling and the AMPK agonist metformin; blockade of HNF4α activity resulted in cyclin downregulation, cell cycle arrest and tumour growth inhibition. HNF4α also regulated WNT signalling through its target gene WNT5A, a potential prognostic marker of diffuse type gastric tumours. CONCLUSIONS Our results indicate that HNF4α is a targetable oncoprotein in GC, is regulated by AMPK signalling through AMPKα and resides upstream of WNT signalling. HNF4α may regulate 'metabolic switch' characteristic of a general malignant phenotype and its target WNT5A has potential prognostic values. The AMPKα-HNF4α-WNT5A signalling cascade represents a potentially targetable pathway for drug development.
Collapse
Affiliation(s)
- Hae Ryung Chang
- New Experimental Therapeutics Branch, National Cancer Center of Korea, Goyang-si, Kyeonggi-do, Republic of Korea
| | - Seungyoon Nam
- New Experimental Therapeutics Branch, National Cancer Center of Korea, Goyang-si, Kyeonggi-do, Republic of Korea
| | - Myeong-Cherl Kook
- Department of Pathology, National Cancer Center of Korea, Goyang-si, Kyeonggi-do, Republic of Korea
| | - Kyung-Tae Kim
- Molecular Epidemiology Branch, National Cancer Center of Korea, Goyang-si, Kyeonggi-do, Republic of Korea
| | - Xiuping Liu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hui Yao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hae Rim Jung
- New Experimental Therapeutics Branch, National Cancer Center of Korea, Goyang-si, Kyeonggi-do, Republic of Korea
| | - Robert Lemos
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California, USA
| | - Hye Hyun Seo
- Animal Sciences Branch, National Cancer Center of Korea, Goyang-si, Kyeonggi-do, Republic of Korea
| | - Hee Seo Park
- New Experimental Therapeutics Branch, National Cancer Center of Korea, Goyang-si, Kyeonggi-do, Republic of Korea
| | - Youme Gim
- New Experimental Therapeutics Branch, National Cancer Center of Korea, Goyang-si, Kyeonggi-do, Republic of Korea
| | - Dongwan Hong
- Cancer Genomics Branch, National Cancer Center of Korea, Goyang-si, Kyeonggi-do, Republic of Korea
| | - Iksoo Huh
- Department of Statistics, Seoul National University, Seoul, Republic of Korea
| | - Young-Woo Kim
- Gastric Cancer Branch, National Cancer Center of Korea, Goyang-si, Kyeonggi-do, Republic of Korea
| | - Dongfeng Tan
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Chang-Gong Liu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Garth Powis
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California, USA
| | - Taesung Park
- Department of Statistics, Seoul National University, Seoul, Republic of Korea
| | - Han Liang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yon Hui Kim
- New Experimental Therapeutics Branch, National Cancer Center of Korea, Goyang-si, Kyeonggi-do, Republic of Korea
| |
Collapse
|
90
|
Popov VB, Jornayvaz FR, Akgul EO, Kanda S, Jurczak MJ, Zhang D, Abudukadier A, Majumdar SK, Guigni B, Petersen KF, Manchem VP, Bhanot S, Shulman GI, Samuel VT. Second-generation antisense oligonucleotides against β-catenin protect mice against diet-induced hepatic steatosis and hepatic and peripheral insulin resistance. FASEB J 2015; 30:1207-17. [PMID: 26644352 DOI: 10.1096/fj.15-271999] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 11/16/2015] [Indexed: 12/24/2022]
Abstract
Although mutations in the Wnt/β-catenin signaling pathway are linked with the metabolic syndrome and type 2 diabetes in humans, the mechanism is unclear. High-fat-fed male C57BL/6 mice were treated for 4 wk with a 2'-O-methoxyethyl chimeric antisense oligonucleotide (ASO) to decrease hepatic and adipose expression of β-catenin. β-Catenin mRNA decreased by ≈80% in the liver and by 70% in white adipose tissue relative to control ASO-treated mice. β-Catenin ASO improved hepatic insulin sensitivity and increased insulin-stimulated whole body glucose metabolism, as assessed during hyperinsulinemic-euglycemic clamp in awake mice. β-Catenin ASO altered hepatic lipid composition in high-fat-fed mice. There were reductions in hepatic triglyceride (44%, P < 0.05) and diacylglycerol content (60%, P < 0.01) but a 30% increase in ceramide content (P < 0.001). The altered lipid content was attributed to decreased expression of sn-1,2 diacylglycerol acyltransferase and mitochondrial acyl-CoA:glycerol-sn-3-phosphate acyltransferase and an increase in serine palmitoyl transferase. The decrease in cellular diacyglycerol was associated with a 33% decrease in PKCε activation (P < 0.05) and 64% increase in Akt2 phosphorylation (P < 0.05). In summary, Reducing β-catenin expression decreases expression of enzymes involved in hepatic fatty acid esterification, ameliorates hepatic steatosis and lipid-induced insulin resistance.
Collapse
Affiliation(s)
- Violeta B Popov
- *Department of Internal Medicine, Department of Cellular and Molecular Physiology, and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA; West Haven Veterans Affairs Medical Center, West Haven, Connecticut, USA; and ISIS Pharmaceuticals, Carlsbad, California, USA
| | - Francois R Jornayvaz
- *Department of Internal Medicine, Department of Cellular and Molecular Physiology, and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA; West Haven Veterans Affairs Medical Center, West Haven, Connecticut, USA; and ISIS Pharmaceuticals, Carlsbad, California, USA
| | - Emin O Akgul
- *Department of Internal Medicine, Department of Cellular and Molecular Physiology, and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA; West Haven Veterans Affairs Medical Center, West Haven, Connecticut, USA; and ISIS Pharmaceuticals, Carlsbad, California, USA
| | - Shoichi Kanda
- *Department of Internal Medicine, Department of Cellular and Molecular Physiology, and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA; West Haven Veterans Affairs Medical Center, West Haven, Connecticut, USA; and ISIS Pharmaceuticals, Carlsbad, California, USA
| | - Michael J Jurczak
- *Department of Internal Medicine, Department of Cellular and Molecular Physiology, and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA; West Haven Veterans Affairs Medical Center, West Haven, Connecticut, USA; and ISIS Pharmaceuticals, Carlsbad, California, USA
| | - Dongyan Zhang
- *Department of Internal Medicine, Department of Cellular and Molecular Physiology, and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA; West Haven Veterans Affairs Medical Center, West Haven, Connecticut, USA; and ISIS Pharmaceuticals, Carlsbad, California, USA
| | - Abulizi Abudukadier
- *Department of Internal Medicine, Department of Cellular and Molecular Physiology, and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA; West Haven Veterans Affairs Medical Center, West Haven, Connecticut, USA; and ISIS Pharmaceuticals, Carlsbad, California, USA
| | - Sachin K Majumdar
- *Department of Internal Medicine, Department of Cellular and Molecular Physiology, and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA; West Haven Veterans Affairs Medical Center, West Haven, Connecticut, USA; and ISIS Pharmaceuticals, Carlsbad, California, USA
| | - Blas Guigni
- *Department of Internal Medicine, Department of Cellular and Molecular Physiology, and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA; West Haven Veterans Affairs Medical Center, West Haven, Connecticut, USA; and ISIS Pharmaceuticals, Carlsbad, California, USA
| | - Kitt Falk Petersen
- *Department of Internal Medicine, Department of Cellular and Molecular Physiology, and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA; West Haven Veterans Affairs Medical Center, West Haven, Connecticut, USA; and ISIS Pharmaceuticals, Carlsbad, California, USA
| | - Vara Prasad Manchem
- *Department of Internal Medicine, Department of Cellular and Molecular Physiology, and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA; West Haven Veterans Affairs Medical Center, West Haven, Connecticut, USA; and ISIS Pharmaceuticals, Carlsbad, California, USA
| | - Sanjay Bhanot
- *Department of Internal Medicine, Department of Cellular and Molecular Physiology, and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA; West Haven Veterans Affairs Medical Center, West Haven, Connecticut, USA; and ISIS Pharmaceuticals, Carlsbad, California, USA
| | - Gerald I Shulman
- *Department of Internal Medicine, Department of Cellular and Molecular Physiology, and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA; West Haven Veterans Affairs Medical Center, West Haven, Connecticut, USA; and ISIS Pharmaceuticals, Carlsbad, California, USA
| | - Varman T Samuel
- *Department of Internal Medicine, Department of Cellular and Molecular Physiology, and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA; West Haven Veterans Affairs Medical Center, West Haven, Connecticut, USA; and ISIS Pharmaceuticals, Carlsbad, California, USA
| |
Collapse
|
91
|
Pigment Epithelium-Derived Factor (PEDF) is a Determinant of Stem Cell Fate: Lessons from an Ultra-Rare Disease. J Dev Biol 2015; 3:112-128. [PMID: 27239449 PMCID: PMC4883593 DOI: 10.3390/jdb3040112] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
PEDF is a secreted glycoprotein that is widely expressed by multiple organs. Numerous functional contributions have been attributed to PEDF with antiangiogenic, antitumor, anti-inflammatory, and neurotrophic properties among the most prominent. The discovery that null mutations in the PEDF gene results in Osteogenesis Imperfecta Type VI, a rare autosomal recessive bone disease characterized by multiple fractures, highlights a critical developmental function for this protein. This ultra-rare orphan disease has provided biological insights into previous studies that noted PEDF’s effects on various stem cell populations. In addition to bone development, PEDF modulates resident stem cell populations in the brain, muscle, and eye. Functional effects on human embryonic stem cells have also been demonstrated. An overview of recent advances in our understanding by which PEDF regulates stem cells and their potential clinical applications will be evaluated in this review.
Collapse
|
92
|
Zhu X, Wu YB, Zhou J, Kang DM. Upregulation of lncRNA MEG3 promotes hepatic insulin resistance via increasing FoxO1 expression. Biochem Biophys Res Commun 2015; 469:319-25. [PMID: 26603935 DOI: 10.1016/j.bbrc.2015.11.048] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 11/10/2015] [Indexed: 01/20/2023]
Abstract
BACKGROUND Hepatic insulin resistance is a major characteristic of type 2 diabetes mellitus. LncRNA MEG3 has been shown to correlate to hepatic glucose production; however, the underlying mechanism remains unclear. This study aims to investigate the role of MEG3 in hepatic insulin resistance. METHODS High-fat diet mice, ob/ob mice and mice primary hepatocytes were used in this study. Expression of MEG3, FoxO1, G6pc and Pepck were determined by real-time PCR. FoxO1, G6pc, Pepck, HDAC1 and HDAC3 protein levels were analyzed by western blotting. Hepatic gluconeogenesis, glycogen accumulation, triglyceride and glycogen contents were measured by corresponding assay or kit, and body weight was monitored after an overnight fast. RESULTS Gene expression of MEG3 was upregulated in high-fat diet and ob/ob mice and increased by palmitate, oleate or linoleate. MEG3 overexpression significantly increased FoxO1, G6pc, Pepck mRNA expressions and hepatic gluconeogenesis and suppressed insulin-stimulated glycogen synthesis in primary hepatocytes, whereas palmitate-induced increase of FoxO1, G6pc and Pepck protein expressions could be reversed by MEG3 interference. In addition, high fat enhanced expression of lncRNA MEG3 in hepatocytes through histone acetylation. Furthermore, MEG3 interference could reverse the up-regulation of triglyceride as well as impaired glucose tolerance and down-regulation of glucogen content in high-fat diet mice or ob/ob mice. CONCLUSION Upregulation of lncRNA MEG3 enhances hepatic insulin resistance via increasing foxO1expression, suggesting that MEG3 may be a potential target and therapeutic strategy for diabetes.
Collapse
Affiliation(s)
- Xiang Zhu
- Department of Gerontology, Affiliated Anhui Provincial Hospital, Anhui Medical University, Hefei, 230001, China.
| | - Yuan-Bo Wu
- Department of Neurology, Affiliated Anhui Provincial Hospital, Anhui Medical University, Hefei, 230001, China
| | - Jian Zhou
- Department of Gerontology, Affiliated Anhui Provincial Hospital, Anhui Medical University, Hefei, 230001, China
| | - Dong-Mei Kang
- Department of Gerontology, Affiliated Anhui Provincial Hospital, Anhui Medical University, Hefei, 230001, China.
| |
Collapse
|
93
|
Current understanding and dispute on the function of the Wnt signaling pathway effector TCF7L2 in hepatic gluconeogenesis. Genes Dis 2015; 3:48-55. [PMID: 30258876 PMCID: PMC6147171 DOI: 10.1016/j.gendis.2015.10.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 10/27/2015] [Indexed: 02/07/2023] Open
Abstract
Approximately 10 years ago, the Wnt signaling pathway effector TCF7L2 (=TCF-4) was recognized as a type 2 diabetes (T2D) risk gene through a genome wide association study (GWAS). As the correlation between TCF7L2 polymorphisms and T2D susceptibility has been reproducibly observed by numerous follow-up investigations among different ethnic groups, great efforts have been made to explore the function of TCF7L2 in metabolic organs including the pancreas, liver and adipose tissues. Although these explorations have enriched our general knowledge on the Wnt signaling cascade in metabolic homeostasis, studies conducted to date have also generated controversial suggestions. Here I will provide a brief review on the Wnt signaling pathway as well as the milestone GWAS discovery and the follow-up studies. I will then discuss the two different opinions on the correlation between TCF7L2 variants and T2D risk, a gain-of-function event versus a loss-of-function event. This will be followed by summarizing the relevant investigations on the metabolic function of hepatic TCF7L2 and presenting our view on the discrepancy and perspectives.
Collapse
|
94
|
Kaur K, Vig S, Srivastava R, Mishra A, Singh VP, Srivastava AK, Datta M. Elevated Hepatic miR-22-3p Expression Impairs Gluconeogenesis by Silencing the Wnt-Responsive Transcription Factor Tcf7. Diabetes 2015; 64:3659-69. [PMID: 26193896 DOI: 10.2337/db14-1924] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 06/29/2015] [Indexed: 11/13/2022]
Abstract
Levels of miR-22-3p, a highly abundant hepatic microRNA, are abnormally increased in mouse models of insulin resistance and type 2 diabetes, yet its contribution to deregulated hepatic metabolism under diseased states is not well understood. Here, we unravel a novel link between elevated hepatic miR-22-3p expression and impaired gluconeogenesis in diabetic db/db mice via the regulation of Tcf7 (transcription factor 7). Our data demonstrate that miR-22-3p binds to the 3' untranslated region of TCF7 and downregulates it, and this microRNA-mediated regulation of TCF7 increases the expression of enzymes of the gluconeogenic pathway in HepG2 cells. Small interfering RNA-mediated knockdown of TCF7 in HepG2 cells also causes similar upregulation of gluconeogenic genes. Furthermore, in vivo silencing of miR-22-3p by antagomiR administration lowered random as well as fasting glucose levels in diabetic mice. miR-22-3p antagonism improved glucose tolerance and insulin sensitivity. Importantly, the hepatic Tcf7 levels were restored along with reduced hepatic glucose output, which was also reflected by the decreased expression of gluconeogenic genes. Our results support a critical role for miR-22-3p and its target, Tcf7, in the pathogenesis of diabetes by upregulating gluconeogenesis. Moreover, targeting the miR-22/Tcf7/Wnt axis might hold therapeutic potential for the treatment of altered hepatic physiology during insulin resistance and type 2 diabetes.
Collapse
Affiliation(s)
- Kirandeep Kaur
- Council of Scientific and Industrial Research-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Saurabh Vig
- Council of Scientific and Industrial Research-Institute of Genomics and Integrative Biology, New Delhi, India Academy of Scientific and Innovative Research, Training and Development Complex, Council of Scientific and Industrial Research Campus, Taramani, Chennai, India
| | - Rohit Srivastava
- Council of Scientific and Industrial Research-Central Drug Research Institute, Jankipuram Extension, Lucknow, India
| | - Akansha Mishra
- Council of Scientific and Industrial Research-Central Drug Research Institute, Jankipuram Extension, Lucknow, India
| | - Vijay Pal Singh
- Council of Scientific and Industrial Research-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Arvind K Srivastava
- Council of Scientific and Industrial Research-Central Drug Research Institute, Jankipuram Extension, Lucknow, India
| | - Malabika Datta
- Council of Scientific and Industrial Research-Institute of Genomics and Integrative Biology, New Delhi, India Academy of Scientific and Innovative Research, Training and Development Complex, Council of Scientific and Industrial Research Campus, Taramani, Chennai, India
| |
Collapse
|
95
|
Daniele G, Winnier D, Mari A, Bruder J, Fourcaudot M, Pengou Z, Tripathy D, Jenkinson C, Folli F. Sclerostin and Insulin Resistance in Prediabetes: Evidence of a Cross Talk Between Bone and Glucose Metabolism. Diabetes Care 2015; 38:1509-17. [PMID: 26084344 DOI: 10.2337/dc14-2989] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 04/28/2015] [Indexed: 02/03/2023]
Abstract
OBJECTIVE A gene mutation of the Wnt/β-catenin signaling cascade is present in rare patients with the insulin resistance syndrome. Sclerostin is a circulating peptide inhibiting Wnt/β-catenin signaling. Our aims were to evaluate serum sclerostin in subjects with prediabetes and to analyze its relationship with insulin resistance and β-cell function. RESEARCH DESIGN AND METHODS We performed a cross-sectional study including 43 healthy normal glucose-tolerant (NGT) individuals and 79 individuals with impaired glucose regulation (IGR), which included subjects with impaired fasting glucose (IFG), impaired glucose tolerance (IGT), and combined IFG-IGT, undergoing oral glucose tolerance test (OGTT) and dual-energy X-ray absorptiometry. A subgroup of 18 with NGT and 30 with IGR also underwent a euglycemic-hyperinsulinemic clamp with tracer. RESULTS Sclerostin levels were higher in IGR compared with NGT (50.8 ± 2.4 vs. 38.7 ± 2.3 pmol/L; P = 0.01), positively correlated with HOMA-insulin resistance (IR) (r = 0.62; P < 0.001), and negatively correlated with insulin-mediated total body glucose disposal (r = -0.40; P < 0.001). Fasting endogenous glucose production (EGP) and hepatic and adipose tissue insulin resistance indexes were positively correlated with sclerostin levels (r = 0.48, r = 0.62, and r = 0.61, respectively; P < 0.001). Fasting and OGTT insulin clearance were inversely correlated with sclerostin serum levels (r = -0.52 and r = -0.44, respectively; both P < 0.001). Sclerostin levels were not correlated with β-cell function parameters. In multiple linear regression analysis, the addition of sclerostin levels to the traditional risk factors for insulin resistance improved the r(2) associated with HOMA-IR (r(2) change: 0.055; F change: 28.893; P = 0.001) and insulin-mediated total body glucose disposal (r(2) change: 0.059; F change: 4.938; P = 0.033). CONCLUSIONS Sclerostin levels are increased in individuals with prediabetes and correlated with insulin resistance in skeletal muscle, liver, and adipose tissue. The correlation between sclerostin and insulin clearance at fasting state and during OGTT is novel; thus, studies are needed to explore the potential causal relationship.
Collapse
Affiliation(s)
- Giuseppe Daniele
- Department of Medicine, Diabetes Division, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Deidre Winnier
- Department of Medicine, Diabetes Division, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Andrea Mari
- Institute of Neuroscience, National Research Council, Padua, Italy
| | - Jan Bruder
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Marcel Fourcaudot
- Department of Medicine, Diabetes Division, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Zuo Pengou
- Department of Medicine, Diabetes Division, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Devjit Tripathy
- Department of Medicine, Diabetes Division, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Christopher Jenkinson
- Department of Medicine, Diabetes Division, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Franco Folli
- Department of Medicine, Diabetes Division, University of Texas Health Science Center at San Antonio, San Antonio, TX Departamento de Clinica Medica, Faculdade de Ciencias Medicas, Obesity and Comorbidities Research Center, Universidade Estadual de Campinas, Campinas, Brazil
| |
Collapse
|
96
|
Okada K, Naito AT, Higo T, Nakagawa A, Shibamoto M, Sakai T, Hashimoto A, Kuramoto Y, Sumida T, Nomura S, Ito M, Yamaguchi T, Oka T, Akazawa H, Lee JK, Morimoto S, Sakata Y, Shiojima I, Komuro I. Wnt/β-Catenin Signaling Contributes to Skeletal Myopathy in Heart Failure via Direct Interaction With Forkhead Box O. Circ Heart Fail 2015; 8:799-808. [DOI: 10.1161/circheartfailure.114.001958] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 05/15/2015] [Indexed: 01/15/2023]
Abstract
Background—
There are changes in the skeletal muscle of patients with chronic heart failure (CHF), such as volume reduction and fiber type shift toward fatigable type IIb fiber. Forkhead box O (FoxO) signaling plays a critical role in the development of skeletal myopathy in CHF, and functional interaction between FoxO and the Wnt signal mediator β-catenin was previously demonstrated. We have recently reported that serum of CHF model mice activates Wnt signaling more potently than serum of control mice and that complement C1q mediates this activation. We, therefore, hypothesized that C1q-induced activation of Wnt signaling plays a critical role in skeletal myopathy via the interaction with FoxO.
Methods and Results—
Fiber type shift toward fatigable fiber was observed in the skeletal muscle of dilated cardiomyopathy model mice, which was associated with activation of both Wnt and FoxO signaling. Wnt3a protein activated FoxO signaling and induced fiber type shift toward fatigable fiber in C2C12 cells. Wnt3a-induced fiber type shift was inhibited by suppression of FoxO1 activity, whereas Wnt3a-independent fiber type shift was observed by overexpression of constitutively active FoxO1. Serum of dilated cardiomyopathy mice activated both Wnt and FoxO signaling and induced fiber type shift toward fatigable fiber in C2C12 cells. Wnt inhibitor and C1-inhibitor attenuated FoxO activation and fiber type shift both in C2C12 cells and in the skeletal muscle of dilated cardiomyopathy mice.
Conclusions—
C1q-induced activation of Wnt signaling contributes to fiber type shift toward fatigable fiber in CHF. Wnt signaling may be a novel therapeutic target to prevent skeletal myopathy in CHF.
Collapse
Affiliation(s)
- Katsuki Okada
- From the Departments of Cardiovascular Medicine (K.O., A.T.N., T.H., A.N., M.S., T.S., A.H., Y.K., T.O., Y.S.) and Cardiovascular Regenerative Medicine (J.-K.L.), Osaka University Graduate School of Medicine, Osaka, Japan; Japan Science and Technology Agency, CREST, Tokyo, Japan (A.T.N., T.S., S.N., T.O., H.A., J.-K.L., I.S., I.K.); Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (A.T.N., T.S., S.N., M.I., T.Y., H.A., I.K.); Department of
| | - Atsuhiko T. Naito
- From the Departments of Cardiovascular Medicine (K.O., A.T.N., T.H., A.N., M.S., T.S., A.H., Y.K., T.O., Y.S.) and Cardiovascular Regenerative Medicine (J.-K.L.), Osaka University Graduate School of Medicine, Osaka, Japan; Japan Science and Technology Agency, CREST, Tokyo, Japan (A.T.N., T.S., S.N., T.O., H.A., J.-K.L., I.S., I.K.); Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (A.T.N., T.S., S.N., M.I., T.Y., H.A., I.K.); Department of
| | - Tomoaki Higo
- From the Departments of Cardiovascular Medicine (K.O., A.T.N., T.H., A.N., M.S., T.S., A.H., Y.K., T.O., Y.S.) and Cardiovascular Regenerative Medicine (J.-K.L.), Osaka University Graduate School of Medicine, Osaka, Japan; Japan Science and Technology Agency, CREST, Tokyo, Japan (A.T.N., T.S., S.N., T.O., H.A., J.-K.L., I.S., I.K.); Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (A.T.N., T.S., S.N., M.I., T.Y., H.A., I.K.); Department of
| | - Akito Nakagawa
- From the Departments of Cardiovascular Medicine (K.O., A.T.N., T.H., A.N., M.S., T.S., A.H., Y.K., T.O., Y.S.) and Cardiovascular Regenerative Medicine (J.-K.L.), Osaka University Graduate School of Medicine, Osaka, Japan; Japan Science and Technology Agency, CREST, Tokyo, Japan (A.T.N., T.S., S.N., T.O., H.A., J.-K.L., I.S., I.K.); Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (A.T.N., T.S., S.N., M.I., T.Y., H.A., I.K.); Department of
| | - Masato Shibamoto
- From the Departments of Cardiovascular Medicine (K.O., A.T.N., T.H., A.N., M.S., T.S., A.H., Y.K., T.O., Y.S.) and Cardiovascular Regenerative Medicine (J.-K.L.), Osaka University Graduate School of Medicine, Osaka, Japan; Japan Science and Technology Agency, CREST, Tokyo, Japan (A.T.N., T.S., S.N., T.O., H.A., J.-K.L., I.S., I.K.); Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (A.T.N., T.S., S.N., M.I., T.Y., H.A., I.K.); Department of
| | - Taku Sakai
- From the Departments of Cardiovascular Medicine (K.O., A.T.N., T.H., A.N., M.S., T.S., A.H., Y.K., T.O., Y.S.) and Cardiovascular Regenerative Medicine (J.-K.L.), Osaka University Graduate School of Medicine, Osaka, Japan; Japan Science and Technology Agency, CREST, Tokyo, Japan (A.T.N., T.S., S.N., T.O., H.A., J.-K.L., I.S., I.K.); Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (A.T.N., T.S., S.N., M.I., T.Y., H.A., I.K.); Department of
| | - Akihito Hashimoto
- From the Departments of Cardiovascular Medicine (K.O., A.T.N., T.H., A.N., M.S., T.S., A.H., Y.K., T.O., Y.S.) and Cardiovascular Regenerative Medicine (J.-K.L.), Osaka University Graduate School of Medicine, Osaka, Japan; Japan Science and Technology Agency, CREST, Tokyo, Japan (A.T.N., T.S., S.N., T.O., H.A., J.-K.L., I.S., I.K.); Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (A.T.N., T.S., S.N., M.I., T.Y., H.A., I.K.); Department of
| | - Yuki Kuramoto
- From the Departments of Cardiovascular Medicine (K.O., A.T.N., T.H., A.N., M.S., T.S., A.H., Y.K., T.O., Y.S.) and Cardiovascular Regenerative Medicine (J.-K.L.), Osaka University Graduate School of Medicine, Osaka, Japan; Japan Science and Technology Agency, CREST, Tokyo, Japan (A.T.N., T.S., S.N., T.O., H.A., J.-K.L., I.S., I.K.); Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (A.T.N., T.S., S.N., M.I., T.Y., H.A., I.K.); Department of
| | - Tomokazu Sumida
- From the Departments of Cardiovascular Medicine (K.O., A.T.N., T.H., A.N., M.S., T.S., A.H., Y.K., T.O., Y.S.) and Cardiovascular Regenerative Medicine (J.-K.L.), Osaka University Graduate School of Medicine, Osaka, Japan; Japan Science and Technology Agency, CREST, Tokyo, Japan (A.T.N., T.S., S.N., T.O., H.A., J.-K.L., I.S., I.K.); Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (A.T.N., T.S., S.N., M.I., T.Y., H.A., I.K.); Department of
| | - Seitaro Nomura
- From the Departments of Cardiovascular Medicine (K.O., A.T.N., T.H., A.N., M.S., T.S., A.H., Y.K., T.O., Y.S.) and Cardiovascular Regenerative Medicine (J.-K.L.), Osaka University Graduate School of Medicine, Osaka, Japan; Japan Science and Technology Agency, CREST, Tokyo, Japan (A.T.N., T.S., S.N., T.O., H.A., J.-K.L., I.S., I.K.); Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (A.T.N., T.S., S.N., M.I., T.Y., H.A., I.K.); Department of
| | - Masamichi Ito
- From the Departments of Cardiovascular Medicine (K.O., A.T.N., T.H., A.N., M.S., T.S., A.H., Y.K., T.O., Y.S.) and Cardiovascular Regenerative Medicine (J.-K.L.), Osaka University Graduate School of Medicine, Osaka, Japan; Japan Science and Technology Agency, CREST, Tokyo, Japan (A.T.N., T.S., S.N., T.O., H.A., J.-K.L., I.S., I.K.); Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (A.T.N., T.S., S.N., M.I., T.Y., H.A., I.K.); Department of
| | - Toshihiro Yamaguchi
- From the Departments of Cardiovascular Medicine (K.O., A.T.N., T.H., A.N., M.S., T.S., A.H., Y.K., T.O., Y.S.) and Cardiovascular Regenerative Medicine (J.-K.L.), Osaka University Graduate School of Medicine, Osaka, Japan; Japan Science and Technology Agency, CREST, Tokyo, Japan (A.T.N., T.S., S.N., T.O., H.A., J.-K.L., I.S., I.K.); Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (A.T.N., T.S., S.N., M.I., T.Y., H.A., I.K.); Department of
| | - Toru Oka
- From the Departments of Cardiovascular Medicine (K.O., A.T.N., T.H., A.N., M.S., T.S., A.H., Y.K., T.O., Y.S.) and Cardiovascular Regenerative Medicine (J.-K.L.), Osaka University Graduate School of Medicine, Osaka, Japan; Japan Science and Technology Agency, CREST, Tokyo, Japan (A.T.N., T.S., S.N., T.O., H.A., J.-K.L., I.S., I.K.); Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (A.T.N., T.S., S.N., M.I., T.Y., H.A., I.K.); Department of
| | - Hiroshi Akazawa
- From the Departments of Cardiovascular Medicine (K.O., A.T.N., T.H., A.N., M.S., T.S., A.H., Y.K., T.O., Y.S.) and Cardiovascular Regenerative Medicine (J.-K.L.), Osaka University Graduate School of Medicine, Osaka, Japan; Japan Science and Technology Agency, CREST, Tokyo, Japan (A.T.N., T.S., S.N., T.O., H.A., J.-K.L., I.S., I.K.); Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (A.T.N., T.S., S.N., M.I., T.Y., H.A., I.K.); Department of
| | - Jong-Kook Lee
- From the Departments of Cardiovascular Medicine (K.O., A.T.N., T.H., A.N., M.S., T.S., A.H., Y.K., T.O., Y.S.) and Cardiovascular Regenerative Medicine (J.-K.L.), Osaka University Graduate School of Medicine, Osaka, Japan; Japan Science and Technology Agency, CREST, Tokyo, Japan (A.T.N., T.S., S.N., T.O., H.A., J.-K.L., I.S., I.K.); Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (A.T.N., T.S., S.N., M.I., T.Y., H.A., I.K.); Department of
| | - Sachio Morimoto
- From the Departments of Cardiovascular Medicine (K.O., A.T.N., T.H., A.N., M.S., T.S., A.H., Y.K., T.O., Y.S.) and Cardiovascular Regenerative Medicine (J.-K.L.), Osaka University Graduate School of Medicine, Osaka, Japan; Japan Science and Technology Agency, CREST, Tokyo, Japan (A.T.N., T.S., S.N., T.O., H.A., J.-K.L., I.S., I.K.); Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (A.T.N., T.S., S.N., M.I., T.Y., H.A., I.K.); Department of
| | - Yasushi Sakata
- From the Departments of Cardiovascular Medicine (K.O., A.T.N., T.H., A.N., M.S., T.S., A.H., Y.K., T.O., Y.S.) and Cardiovascular Regenerative Medicine (J.-K.L.), Osaka University Graduate School of Medicine, Osaka, Japan; Japan Science and Technology Agency, CREST, Tokyo, Japan (A.T.N., T.S., S.N., T.O., H.A., J.-K.L., I.S., I.K.); Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (A.T.N., T.S., S.N., M.I., T.Y., H.A., I.K.); Department of
| | - Ichiro Shiojima
- From the Departments of Cardiovascular Medicine (K.O., A.T.N., T.H., A.N., M.S., T.S., A.H., Y.K., T.O., Y.S.) and Cardiovascular Regenerative Medicine (J.-K.L.), Osaka University Graduate School of Medicine, Osaka, Japan; Japan Science and Technology Agency, CREST, Tokyo, Japan (A.T.N., T.S., S.N., T.O., H.A., J.-K.L., I.S., I.K.); Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (A.T.N., T.S., S.N., M.I., T.Y., H.A., I.K.); Department of
| | - Issei Komuro
- From the Departments of Cardiovascular Medicine (K.O., A.T.N., T.H., A.N., M.S., T.S., A.H., Y.K., T.O., Y.S.) and Cardiovascular Regenerative Medicine (J.-K.L.), Osaka University Graduate School of Medicine, Osaka, Japan; Japan Science and Technology Agency, CREST, Tokyo, Japan (A.T.N., T.S., S.N., T.O., H.A., J.-K.L., I.S., I.K.); Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (A.T.N., T.S., S.N., M.I., T.Y., H.A., I.K.); Department of
| |
Collapse
|
97
|
Monga SP. β-Catenin Signaling and Roles in Liver Homeostasis, Injury, and Tumorigenesis. Gastroenterology 2015; 148:1294-310. [PMID: 25747274 PMCID: PMC4494085 DOI: 10.1053/j.gastro.2015.02.056] [Citation(s) in RCA: 498] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 02/21/2015] [Accepted: 02/23/2015] [Indexed: 12/11/2022]
Abstract
β-catenin (encoded by CTNNB1) is a subunit of the cell surface cadherin protein complex that acts as an intracellular signal transducer in the WNT signaling pathway; alterations in its activity have been associated with the development of hepatocellular carcinoma and other liver diseases. Other than WNT, additional signaling pathways also can converge at β-catenin. β-catenin also interacts with transcription factors such as T-cell factor, forkhead box protein O, and hypoxia inducible factor 1α to regulate the expression of target genes. We discuss the role of β-catenin in metabolic zonation of the adult liver. β-catenin also regulates the expression of genes that control metabolism of glucose, nutrients, and xenobiotics; alterations in its activity may contribute to the pathogenesis of nonalcoholic steatohepatitis. Alterations in β-catenin signaling may lead to activation of hepatic stellate cells, which is required for fibrosis. Many hepatic tumors such as hepatocellular adenomas, hepatocellular cancers, and hepatoblastomas have mutations in CTNNB1 that result in constitutive activation of β-catenin, so this molecule could be a therapeutic target. We discuss how alterations in β-catenin activity contribute to liver disease and how these might be used in diagnosis and prognosis, as well as in the development of therapeutics.
Collapse
Affiliation(s)
- Satdarshan Pal Monga
- Department of Pathology and Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| |
Collapse
|
98
|
Ip W, Shao W, Song Z, Chen Z, Wheeler MB, Jin T. Liver-specific expression of dominant-negative transcription factor 7-like 2 causes progressive impairment in glucose homeostasis. Diabetes 2015; 64:1923-32. [PMID: 25576056 DOI: 10.2337/db14-1329] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 01/01/2015] [Indexed: 11/13/2022]
Abstract
Investigations on the metabolic role of the Wnt signaling pathway and hepatic transcription factor 7-like 2 (TCF7L2) have generated opposing views. While some studies demonstrated a repressive effect of TCF7L2 on hepatic gluconeogenesis, a recent study using liver-specific Tcf7l2(-/-) mice suggested the opposite. As a consequence of redundant and bidirectional actions of transcription factor (TCF) molecules and other complexities of the Wnt pathway, knockout of a single Wnt pathway component may not effectively reveal a complete metabolic picture of this pathway. To address this, we generated the liver-specific dominant-negative (DN) TCF7L2 (TCF7L2DN) transgenic mouse model LTCFDN. These mice exhibited progressive impairment in response to pyruvate challenge. Importantly, LTCFDN hepatocytes displayed elevated gluconeogenic gene expression, gluconeogenesis, and loss of Wnt-3a-mediated repression of gluconeogenesis. In C57BL/6 hepatocytes, adenovirus-mediated expression of TCF7L2DN, but not wild-type TCF7L2, increased gluconeogenesis and gluconeogenic gene expression. Our further mechanistic exploration suggests that TCF7L2DN-mediated inhibition of Wnt signaling causes preferential interaction of β-catenin (β-cat) with FoxO1 and increased binding of β-cat/FoxO1 to the Pck1 FoxO binding site, resulting in the stimulation of Pck1 expression and increased gluconeogenesis. Together, our results using TCF7L2DN as a unique tool revealed that the Wnt signaling pathway and its effector β-cat/TCF serve a beneficial role in suppressing hepatic gluconeogenesis.
Collapse
Affiliation(s)
- Wilfred Ip
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Weijuan Shao
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Zhuolun Song
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Zonglan Chen
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Michael B Wheeler
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Tianru Jin
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
99
|
Schleicher J, Tokarski C, Marbach E, Matz-Soja M, Zellmer S, Gebhardt R, Schuster S. Zonation of hepatic fatty acid metabolism - The diversity of its regulation and the benefit of modeling. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:641-56. [PMID: 25677822 DOI: 10.1016/j.bbalip.2015.02.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 01/26/2015] [Accepted: 02/03/2015] [Indexed: 02/07/2023]
Abstract
A pronounced heterogeneity between hepatocytes in subcellular structure and enzyme activities was discovered more than 50years ago and initiated the idea of metabolic zonation. In the last decades zonation patterns of liver metabolism were extensively investigated for carbohydrate, nitrogen and lipid metabolism. The present review focuses on zonation patterns of the latter. We review recent findings regarding the zonation of fatty acid uptake and oxidation, ketogenesis, triglyceride synthesis and secretion, de novo lipogenesis, as well as bile acid and cholesterol metabolism. In doing so, we expose knowledge gaps and discuss contradictory experimental results, for example on the zonation pattern of fatty acid oxidation and de novo lipogenesis. Thus, possible rewarding directions of further research are identified. Furthermore, recent findings about the regulation of metabolic zonation are summarized, especially regarding the role of hormones, nerve innervation, morphogens, gender differences and the influence of the circadian clock. In the last part of the review, a short collection of models considering hepatic lipid metabolism is provided. We conclude that modeling, despite its proven benefit for understanding of hepatic carbohydrate and ammonia metabolisms, has so far been largely disregarded in the study of lipid metabolism; therefore some possible fields of modeling interest are presented.
Collapse
Affiliation(s)
- J Schleicher
- Department of Bioinformatics, University of Jena, Jena, Germany.
| | - C Tokarski
- Department of Bioinformatics, University of Jena, Jena, Germany
| | - E Marbach
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - M Matz-Soja
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - S Zellmer
- Department of Chemicals and Product Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - R Gebhardt
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - S Schuster
- Department of Bioinformatics, University of Jena, Jena, Germany
| |
Collapse
|
100
|
Abstract
The liver is an essential metabolic organ, and its metabolic function is controlled by insulin and other metabolic hormones. Glucose is converted into pyruvate through glycolysis in the cytoplasm, and pyruvate is subsequently oxidized in the mitochondria to generate ATP through the TCA cycle and oxidative phosphorylation. In the fed state, glycolytic products are used to synthesize fatty acids through de novo lipogenesis. Long-chain fatty acids are incorporated into triacylglycerol, phospholipids, and/or cholesterol esters in hepatocytes. These complex lipids are stored in lipid droplets and membrane structures, or secreted into the circulation as very low-density lipoprotein particles. In the fasted state, the liver secretes glucose through both glycogenolysis and gluconeogenesis. During pronged fasting, hepatic gluconeogenesis is the primary source for endogenous glucose production. Fasting also promotes lipolysis in adipose tissue, resulting in release of nonesterified fatty acids which are converted into ketone bodies in hepatic mitochondria though β-oxidation and ketogenesis. Ketone bodies provide a metabolic fuel for extrahepatic tissues. Liver energy metabolism is tightly regulated by neuronal and hormonal signals. The sympathetic system stimulates, whereas the parasympathetic system suppresses, hepatic gluconeogenesis. Insulin stimulates glycolysis and lipogenesis but suppresses gluconeogenesis, and glucagon counteracts insulin action. Numerous transcription factors and coactivators, including CREB, FOXO1, ChREBP, SREBP, PGC-1α, and CRTC2, control the expression of the enzymes which catalyze key steps of metabolic pathways, thus controlling liver energy metabolism. Aberrant energy metabolism in the liver promotes insulin resistance, diabetes, and nonalcoholic fatty liver diseases.
Collapse
Affiliation(s)
- Liangyou Rui
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|