1
|
Moorwood K, Smith FM, Garfield AS, Cowley M, Holt LJ, Daly RJ, Ward A. Grb7, Grb10 and Grb14, encoding the growth factor receptor-bound 7 family of signalling adaptor proteins have overlapping functions in the regulation of fetal growth and post-natal glucose metabolism. BMC Biol 2024; 22:221. [PMID: 39343875 PMCID: PMC11441139 DOI: 10.1186/s12915-024-02018-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND The growth factor receptor bound protein 7 (Grb7) family of signalling adaptor proteins comprises Grb7, Grb10 and Grb14. Each can interact with the insulin receptor and other receptor tyrosine kinases, where Grb10 and Grb14 inhibit insulin receptor activity. In cell culture studies they mediate functions including cell survival, proliferation, and migration. Mouse knockout (KO) studies have revealed physiological roles for Grb10 and Grb14 in glucose-regulated energy homeostasis. Both Grb10 KO and Grb14 KO mice exhibit increased insulin signalling in peripheral tissues, with increased glucose and insulin sensitivity and a modestly increased ability to clear a glucose load. In addition, Grb10 strongly inhibits fetal growth such that at birth Grb10 KO mice are 30% larger by weight than wild type littermates. RESULTS Here, we generate a Grb7 KO mouse model. We show that during fetal development the expression patterns of Grb7 and Grb14 each overlap with that of Grb10. Despite this, Grb7 and Grb14 did not have a major role in influencing fetal growth, either alone or in combination with Grb10. At birth, in most respects both Grb7 KO and Grb14 KO single mutants were indistinguishable from wild type, while Grb7:Grb10 double knockout (DKO) were near identical to Grb10 KO single mutants and Grb10:Grb14 DKO mutants were slightly smaller than Grb10 KO single mutants. In the developing kidney Grb7 had a subtle positive influence on growth. An initial characterisation of Grb7 KO adult mice revealed sexually dimorphic effects on energy homeostasis, with females having a significantly smaller renal white adipose tissue depot and an enhanced ability to clear glucose from the circulation, compared to wild type littermates. Males had elevated fasted glucose levels with a trend towards smaller white adipose depots, without improved glucose clearance. CONCLUSIONS Grb7 and Grb14 do not have significant roles as inhibitors of fetal growth, unlike Grb10, and instead Grb7 may promote growth of the developing kidney. In adulthood, Grb7 contributes subtly to glucose mediated energy homeostasis, raising the possibility of redundancy between all three adaptors in physiological regulation of insulin signalling and glucose handling.
Collapse
Affiliation(s)
- Kim Moorwood
- Department of Life Sciences, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Florentia M Smith
- Department of Life Sciences, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Alastair S Garfield
- Department of Life Sciences, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Michael Cowley
- Department of Life Sciences, University of Bath, Claverton Down, Bath, BA2 7AY, UK
- Present Address: Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University, Campus, Box 7633, Raleigh, NC, 27695, USA
| | - Lowenna J Holt
- Cancer Research Program, Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, NSW, 2010, Australia
| | - Roger J Daly
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Andrew Ward
- Department of Life Sciences, University of Bath, Claverton Down, Bath, BA2 7AY, UK.
| |
Collapse
|
2
|
Das SS, Das SK. Common and ethnic-specific derangements in skeletal muscle transcriptome associated with obesity. Int J Obes (Lond) 2024; 48:330-338. [PMID: 37993634 DOI: 10.1038/s41366-023-01417-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/25/2023] [Accepted: 11/07/2023] [Indexed: 11/24/2023]
Abstract
BACKGROUND Obesity is a common disease with a higher prevalence among African Americans. Obesity alters cellular function in many tissues, including skeletal muscle, and is a risk factor for many life-threatening diseases, including cardiovascular disease and diabetes. The similarities and differences in molecular mechanisms that may explain ethnic disparities in obesity between African and European ancestry individuals have not been studied. METHODS In this study, data from transcriptome-wide analyses on skeletal muscle tissues from well-powered human cohorts were used to compare genes and biological pathways affected by obesity in European and African ancestry populations. Data on obesity-induced differentially expressed transcripts and GWAS-identified SNPs were integrated to prioritize target genes for obesity-associated genetic variants. RESULTS Linear regression analysis in the FUSION (European, N = 301) and AAGMEx (African American, N = 256) cohorts identified a total of 2569 body mass index (BMI)-associated transcripts (q < 0.05), of which 970 genes (at p < 0.05) are associated in both cohorts, and the majority showed the same direction of effect on BMI. Biological pathway analyses, including over-representation and gene-set enrichment analyses, identified enrichment of protein synthesis pathways (e.g., ribosomal function) and the ceramide signaling pathway in both cohorts among BMI-associated down- and up-regulated transcripts, respectively. A comparison using the IPA-tool suggested the activation of inflammation pathways only in Europeans with obesity. Interestingly, these analyses suggested repression of the mitochondrial oxidative phosphorylation pathway in Europeans but showed its activation in African Americans. Integration of SNP-to-Gene analyses-predicted target genes for obesity-associated genetic variants (GWAS-identified SNPs) and BMI-associated transcripts suggested that these SNPs might cause obesity by altering the expression of 316 critical target genes (e.g., GRB14) in the muscle. CONCLUSIONS This study provides a replication of obesity-associated transcripts and biological pathways in skeletal muscle across ethnicities, but also identifies obesity-associated processes unique in either African or European ancestry populations.
Collapse
Affiliation(s)
- Sreejon S Das
- The School of Biotechnology at Atkins, Atkins Academic and Technology High, Winston-Salem, NC, 27101, USA
| | - Swapan K Das
- Department of Internal Medicine, Section of Endocrinology and Metabolism, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
3
|
Ochi Y, Matsui T, Inoue K, Monobe K, Sakamoto H, Aoki S, Taira J. Computational Screening and Experimental Validation of Inhibitor Targeting the Complex Formation of Grb14 and Insulin Receptor. Molecules 2023; 29:198. [PMID: 38202781 PMCID: PMC10780909 DOI: 10.3390/molecules29010198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 12/25/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
The development of drugs targeting gene products associated with insulin resistance holds the potential to enhance our understanding of type 2 diabetes mellitus (T2DM). The virtual screening, based on a three-dimensional (3D) protein structure, is a potential technique to accelerate the development of molecular target drugs. Among the targets implicated in insulin resistance, the genetic characterization and protein function of Grb14 have been clarified without contradiction. The Grb14 gene displays significant variations in T2DM, and its gene product is known to inhibit the function of the insulin receptor (IR) by directly binding to the tyrosine kinase domain. In the present study, a virtual screening, based on a 3D structure of the IR tyrosine kinase domain (IRβ) in complex with part of Grb14, was conducted to find compounds that can disrupt the complex formation between Grb14 and IRβ. First, ten compounds were selected from 154,118 compounds via hierarchical in silico structure-based drug screening, composed of grid docking-based and genetic algorithm-based programs. The experimental validations suggested that the one compound can affect the blood glucose level. The molecular dynamics simulations and co-immunoprecipitation analysis showed that the compound did not completely suppress the protein-protein interaction between Grb14 and IR, though competitively bound to IR with the tyrosine kinase pseudosubstrate region in Grb14.
Collapse
Affiliation(s)
- Yosuke Ochi
- Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, Iizuka 820-8502, Japan
| | - Takanori Matsui
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Keitaro Inoue
- Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, Iizuka 820-8502, Japan
| | - Kohei Monobe
- Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, Iizuka 820-8502, Japan
| | - Hiroshi Sakamoto
- Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, Iizuka 820-8502, Japan
| | - Shunsuke Aoki
- Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, Iizuka 820-8502, Japan
| | - Junichi Taira
- Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, Iizuka 820-8502, Japan
| |
Collapse
|
4
|
Abstract
The insulin receptor (IR) is a type II receptor tyrosine kinase that plays essential roles in metabolism, growth, and proliferation. Dysregulation of IR signaling is linked to many human diseases, such as diabetes and cancers. The resolution revolution in cryo-electron microscopy has led to the determination of several structures of IR with different numbers of bound insulin molecules in recent years, which have tremendously improved our understanding of how IR is activated by insulin. Here, we review the insulin-induced activation mechanism of IR, including (a) the detailed binding modes and functions of insulin at site 1 and site 2 and (b) the insulin-induced structural transitions that are required for IR activation. We highlight several other key aspects of the activation and regulation of IR signaling and discuss the remaining gaps in our understanding of the IR activation mechanism and potential avenues of future research.
Collapse
Affiliation(s)
- Eunhee Choi
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA;
| | - Xiao-Chen Bai
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas, USA;
| |
Collapse
|
5
|
Glunk V, Laber S, Sinnott-Armstrong N, Sobreira DR, Strobel SM, Batista TM, Kubitz P, Moud BN, Ebert H, Huang Y, Brandl B, Garbo G, Honecker J, Stirling DR, Abdennur N, Calabuig-Navarro V, Skurk T, Ocvirk S, Stemmer K, Cimini BA, Carpenter AE, Dankel SN, Lindgren CM, Hauner H, Nobrega MA, Claussnitzer M. A non-coding variant linked to metabolic obesity with normal weight affects actin remodelling in subcutaneous adipocytes. Nat Metab 2023; 5:861-879. [PMID: 37253881 PMCID: PMC11533588 DOI: 10.1038/s42255-023-00807-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 04/12/2023] [Indexed: 06/01/2023]
Abstract
Recent large-scale genomic association studies found evidence for a genetic link between increased risk of type 2 diabetes and decreased risk for adiposity-related traits, reminiscent of metabolically obese normal weight (MONW) association signatures. However, the target genes and cellular mechanisms driving such MONW associations remain to be identified. Here, we systematically identify the cellular programmes of one of the top-scoring MONW risk loci, the 2q24.3 risk locus, in subcutaneous adipocytes. We identify a causal genetic variant, rs6712203, an intronic single-nucleotide polymorphism in the COBLL1 gene, which changes the conserved transcription factor motif of POU domain, class 2, transcription factor 2, and leads to differential COBLL1 gene expression by altering the enhancer activity at the locus in subcutaneous adipocytes. We then establish the cellular programme under the genetic control of the 2q24.3 MONW risk locus and the effector gene COBLL1, which is characterized by impaired actin cytoskeleton remodelling in differentiating subcutaneous adipocytes and subsequent failure of these cells to accumulate lipids and develop into metabolically active and insulin-sensitive adipocytes. Finally, we show that perturbations of the effector gene Cobll1 in a mouse model result in organismal phenotypes matching the MONW association signature, including decreased subcutaneous body fat mass and body weight along with impaired glucose tolerance. Taken together, our results provide a mechanistic link between the genetic risk for insulin resistance and low adiposity, providing a potential therapeutic hypothesis and a framework for future identification of causal relationships between genome associations and cellular programmes in other disorders.
Collapse
Affiliation(s)
- Viktoria Glunk
- Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, Munich, Germany
- ZIEL Institute for Food & Health, Else Kröner-Fresenius-Center for Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Samantha Laber
- Broad Institute of MIT and Harvard, Medical and Population Genetics Program & Type 2 Diabetes Systems Genomics Initiative, Cambridge, MA, USA
| | - Nasa Sinnott-Armstrong
- Broad Institute of MIT and Harvard, Medical and Population Genetics Program & Type 2 Diabetes Systems Genomics Initiative, Cambridge, MA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Herbold Computational Biology Program, Publich Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Debora R Sobreira
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Sophie M Strobel
- Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, Munich, Germany
- ZIEL Institute for Food & Health, Else Kröner-Fresenius-Center for Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising, Germany
- Broad Institute of MIT and Harvard, Medical and Population Genetics Program & Type 2 Diabetes Systems Genomics Initiative, Cambridge, MA, USA
| | - Thiago M Batista
- Broad Institute of MIT and Harvard, Medical and Population Genetics Program & Type 2 Diabetes Systems Genomics Initiative, Cambridge, MA, USA
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Phil Kubitz
- Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, Munich, Germany
- ZIEL Institute for Food & Health, Else Kröner-Fresenius-Center for Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising, Germany
- Broad Institute of MIT and Harvard, Medical and Population Genetics Program & Type 2 Diabetes Systems Genomics Initiative, Cambridge, MA, USA
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bahareh Nemati Moud
- Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, Munich, Germany
- ZIEL Institute for Food & Health, Else Kröner-Fresenius-Center for Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Hannah Ebert
- Institute of Nutritional Sciences, University of Hohenheim, Stuttgart, Germany
| | - Yi Huang
- Broad Institute of MIT and Harvard, Medical and Population Genetics Program & Type 2 Diabetes Systems Genomics Initiative, Cambridge, MA, USA
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Beate Brandl
- Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, Munich, Germany
- ZIEL Institute for Food & Health, Else Kröner-Fresenius-Center for Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Garrett Garbo
- Broad Institute of MIT and Harvard, Medical and Population Genetics Program & Type 2 Diabetes Systems Genomics Initiative, Cambridge, MA, USA
| | - Julius Honecker
- Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, Munich, Germany
- ZIEL Institute for Food & Health, Else Kröner-Fresenius-Center for Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - David R Stirling
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nezar Abdennur
- Institute for Medical Engineering and Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Virtu Calabuig-Navarro
- Broad Institute of MIT and Harvard, Medical and Population Genetics Program & Type 2 Diabetes Systems Genomics Initiative, Cambridge, MA, USA
- Institute of Nutritional Sciences, University of Hohenheim, Stuttgart, Germany
| | - Thomas Skurk
- Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, Munich, Germany
- ZIEL Institute for Food & Health, Else Kröner-Fresenius-Center for Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Soeren Ocvirk
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Intestinal Microbiology Research Group, Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Kerstin Stemmer
- Molecular Cell Biology, Institute for Theoretical Medicine, University of Augsburg, Augsburg, Germany
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Beth A Cimini
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Anne E Carpenter
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Simon N Dankel
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Cecilia M Lindgren
- Broad Institute of MIT and Harvard, Medical and Population Genetics Program & Type 2 Diabetes Systems Genomics Initiative, Cambridge, MA, USA
- Big Data Institute at the Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Hans Hauner
- Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, Munich, Germany
- ZIEL Institute for Food & Health, Else Kröner-Fresenius-Center for Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Marcelo A Nobrega
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Melina Claussnitzer
- Broad Institute of MIT and Harvard, Medical and Population Genetics Program & Type 2 Diabetes Systems Genomics Initiative, Cambridge, MA, USA.
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Sun C, Förster F, Gutsmann B, Moulla Y, Stroh C, Dietrich A, Schön MR, Gärtner D, Lohmann T, Dressler M, Stumvoll M, Blüher M, Kovacs P, Breitfeld J, Guiu-Jurado E. Metabolic Effects of the Waist-To-Hip Ratio Associated Locus GRB14/COBLL1 Are Related to GRB14 Expression in Adipose Tissue. Int J Mol Sci 2022; 23:ijms23158558. [PMID: 35955692 PMCID: PMC9369072 DOI: 10.3390/ijms23158558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 02/04/2023] Open
Abstract
GRB14/COBLL1 locus has been shown to be associated with body fat distribution (FD), but neither the causal gene nor its role in metabolic diseases has been elucidated. We hypothesize that GRB14/COBLL1 may act as the causal genes for FD-related SNPs (rs10195252 and rs6738627), and that they may be regulated by SNP to effect obesity-related metabolic traits. We genotyped rs10195252 and rs6738627 in 2860 subjects with metabolic phenotypes. In a subgroup of 560 subjects, we analyzed GRB14/COBLL1 gene expression in paired visceral and subcutaneous adipose tissue (AT) samples. Mediation analyses were used to determine the causal relationship between SNPs, AT GRB14/COBLL1 mRNA expression, and obesity-related traits. In vitro gene knockdown of Grb14/Cobll1 was used to test their role in adipogenesis. Both gene expressions in AT are correlated with waist circumference. Visceral GRB14 mRNA expression is associated with FPG and HbA1c. Both SNPs are associated with triglycerides, FPG, and leptin levels. Rs10195252 is associated with HbA1c and seems to be mediated by visceral AT GRB14 mRNA expression. Our data support the role of the GRB14/COBLL1 gene expression in body FD and its locus in metabolic sequelae: in particular, lipid metabolism and glucose homeostasis, which is likely mediated by AT GRB14 transcript levels.
Collapse
Affiliation(s)
- Chang Sun
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; (C.S.); (F.F.); (B.G.); (M.S.); (M.B.); (P.K.); (J.B.)
| | - Franz Förster
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; (C.S.); (F.F.); (B.G.); (M.S.); (M.B.); (P.K.); (J.B.)
| | - Beate Gutsmann
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; (C.S.); (F.F.); (B.G.); (M.S.); (M.B.); (P.K.); (J.B.)
| | - Yusef Moulla
- Clinic for Visceral, Transplantation and Thorax and Vascular Surgery, University Hospital Leipzig, 04103 Leipzig, Germany; (Y.M.); (A.D.)
| | - Christine Stroh
- Departement of Obesity and Metabolic Surgery, SRH Wald-Klinikum Gera Str.d. Friedens 122, 07548 Gera, Germany;
| | - Arne Dietrich
- Clinic for Visceral, Transplantation and Thorax and Vascular Surgery, University Hospital Leipzig, 04103 Leipzig, Germany; (Y.M.); (A.D.)
| | - Michael R. Schön
- Städtisches Klinikum Karlsruhe, Clinic of Visceral Surgery, 76133 Karlsruhe, Germany; (M.R.S.); (D.G.)
| | - Daniel Gärtner
- Städtisches Klinikum Karlsruhe, Clinic of Visceral Surgery, 76133 Karlsruhe, Germany; (M.R.S.); (D.G.)
| | - Tobias Lohmann
- Municipal Clinic Dresden-Neustadt, 01129 Dresden, Germany; (T.L.); (M.D.)
| | - Miriam Dressler
- Municipal Clinic Dresden-Neustadt, 01129 Dresden, Germany; (T.L.); (M.D.)
| | - Michael Stumvoll
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; (C.S.); (F.F.); (B.G.); (M.S.); (M.B.); (P.K.); (J.B.)
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Matthias Blüher
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; (C.S.); (F.F.); (B.G.); (M.S.); (M.B.); (P.K.); (J.B.)
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Peter Kovacs
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; (C.S.); (F.F.); (B.G.); (M.S.); (M.B.); (P.K.); (J.B.)
| | - Jana Breitfeld
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; (C.S.); (F.F.); (B.G.); (M.S.); (M.B.); (P.K.); (J.B.)
| | - Esther Guiu-Jurado
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; (C.S.); (F.F.); (B.G.); (M.S.); (M.B.); (P.K.); (J.B.)
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
- Deutsches Zentrum für Diabetesforschung e.V., 85764 Neuherberg, Germany
- Correspondence:
| |
Collapse
|
7
|
Lin CC, Suen KM, Jeffrey PA, Wieteska L, Lidster JA, Bao P, Curd AP, Stainthorp A, Seiler C, Koss H, Miska E, Ahmed Z, Evans SD, Molina-París C, Ladbury JE. Receptor tyrosine kinases regulate signal transduction through a liquid-liquid phase separated state. Mol Cell 2022; 82:1089-1106.e12. [PMID: 35231400 PMCID: PMC8937303 DOI: 10.1016/j.molcel.2022.02.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/02/2021] [Accepted: 02/01/2022] [Indexed: 11/20/2022]
Abstract
The recruitment of signaling proteins into activated receptor tyrosine kinases (RTKs) to produce rapid, high-fidelity downstream response is exposed to the ambiguity of random diffusion to the target site. Liquid-liquid phase separation (LLPS) overcomes this by providing elevated, localized concentrations of the required proteins while impeding competitor ligands. Here, we show a subset of phosphorylation-dependent RTK-mediated LLPS states. We then investigate the formation of phase-separated droplets comprising a ternary complex including the RTK, (FGFR2); the phosphatase, SHP2; and the phospholipase, PLCγ1, which assembles in response to receptor phosphorylation. SHP2 and activated PLCγ1 interact through their tandem SH2 domains via a previously undescribed interface. The complex of FGFR2 and SHP2 combines kinase and phosphatase activities to control the phosphorylation state of the assembly while providing a scaffold for active PLCγ1 to facilitate access to its plasma membrane substrate. Thus, LLPS modulates RTK signaling, with potential consequences for therapeutic intervention.
Collapse
Affiliation(s)
- Chi-Chuan Lin
- School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK.
| | - Kin Man Suen
- School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK; Wellcome Trust Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | | | - Lukasz Wieteska
- School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Jessica A Lidster
- School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Peng Bao
- School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, UK
| | - Alistair P Curd
- School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Amy Stainthorp
- School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Caroline Seiler
- School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Hans Koss
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK; Francis Crick Institute, London NW1 1AT, UK
| | - Eric Miska
- Wellcome Trust Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Zamal Ahmed
- Department of Molecular and Cellular Oncology, University of Texas M D Anderson Cancer Center, Houston, TX 77030, USA
| | - Stephen D Evans
- School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, UK
| | | | - John E Ladbury
- School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
8
|
Kumar L, Vizgaudis W, Klein-Seetharaman J. Structure-based survey of ligand binding in the human insulin receptor. Br J Pharmacol 2021; 179:3512-3528. [PMID: 34907529 DOI: 10.1111/bph.15777] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 11/17/2021] [Accepted: 11/25/2021] [Indexed: 11/27/2022] Open
Abstract
The insulin receptor is a membrane protein responsible for regulation of nutrient balance and therefore an attractive target in the treatment of diabetes and metabolic syndrome. Pharmacology of the insulin receptor involves two distinct mechanisms, (1) activation of the receptor by insulin mimetics that bind in the extracellular domain and (2) inhibition of the receptor tyrosine kinase enzymatic activity in the cytoplasmic domain. While a complete structural picture of the full-length receptor comprising the entire sequence covering extracellular, transmembrane, juxtamembrane and cytoplasmic domains is still elusive, recent progress through cryoelectron microscopy has made it possible to describe the initial insulin ligand binding events at atomistic detail. We utilize this opportunity to obtain structural insights into the pharmacology of the insulin receptor. To this end, we conducted a comprehensive docking study of known ligands to the new structures of the receptor. Through this approach, we provide an in-depth, structure-based review of human insulin receptor pharmacology in light of the new structures.
Collapse
Affiliation(s)
- Lokender Kumar
- Department of Physics, Colorado School of Mines, Golden, CO
| | | | - Judith Klein-Seetharaman
- Department of Chemistry, Colorado School of Mines, Golden, CO.,School of Molecular Sciences & College of Health Solutions, Arizona State University, Phoenix, AZ
| |
Collapse
|
9
|
Rajala RVS, McCauley A, Rajala R, Teel K, Rajala A. Regulation of Phosphoinositide Levels in the Retina by Protein Tyrosine Phosphatase 1B and Growth Factor Receptor-Bound Protein 14. Biomolecules 2021; 11:biom11040602. [PMID: 33921658 PMCID: PMC8073254 DOI: 10.3390/biom11040602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/09/2021] [Accepted: 04/15/2021] [Indexed: 11/16/2022] Open
Abstract
Protein tyrosine kinases and protein phosphatases play a critical role in cellular regulation. The length of a cellular response depends on the interplay between activating protein kinases and deactivating protein phosphatases. Protein tyrosine phosphatase 1B (PTP1B) and growth factor receptor-bound protein 14 (Grb14) are negative regulators of receptor tyrosine kinases. However, in the retina, we have previously shown that PTP1B inactivates insulin receptor signaling, whereas phosphorylated Grb14 inhibits PTP1B activity. In silico docking of phosphorylated Grb14 and PTP1B indicate critical residues in PTP1B that may mediate the interaction. Phosphoinositides (PIPs) are acidic lipids and minor constituents in the cell that play an important role in cellular processes. Their levels are regulated by growth factor signaling. Using phosphoinositide binding protein probes, we observed increased levels of PI(3)P, PI(4)P, PI(3,4)P2, PI(4,5)P2, and PI(3,4,5)P3 in PTP1B knockout mouse retina and decreased levels of these PIPs in Grb14 knockout mouse retina. These observations suggest that the interplay between PTP1B and Grb14 can regulate PIP metabolism.
Collapse
Affiliation(s)
- Raju V. S. Rajala
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (A.M.); (K.T.); (A.R.)
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Dean McGee Eye Institute, Oklahoma City, OK 73104, USA
- Correspondence: ; Tel.: +1-405-271-8255; Fax: +1-405-271-8128
| | - Austin McCauley
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (A.M.); (K.T.); (A.R.)
- Dean McGee Eye Institute, Oklahoma City, OK 73104, USA
| | - Rahul Rajala
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Cardiovascular Biology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Kenneth Teel
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (A.M.); (K.T.); (A.R.)
- Dean McGee Eye Institute, Oklahoma City, OK 73104, USA
| | - Ammaji Rajala
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (A.M.); (K.T.); (A.R.)
- Dean McGee Eye Institute, Oklahoma City, OK 73104, USA
| |
Collapse
|
10
|
Luo P, Zheng M, Zhang R, Zhang H, Liu Y, Li W, Sun X, Yu Q, Tipoe GL, Xiao J. S-Allylmercaptocysteine improves alcoholic liver disease partly through a direct modulation of insulin receptor signaling. Acta Pharm Sin B 2021; 11:668-679. [PMID: 33777674 PMCID: PMC7982498 DOI: 10.1016/j.apsb.2020.11.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/31/2020] [Accepted: 09/07/2020] [Indexed: 12/18/2022] Open
Abstract
Alcoholic liver disease (ALD) causes insulin resistance, lipid metabolism dysfunction, and inflammation. We investigated the protective effects and direct regulating target of S-allylmercaptocysteine (SAMC) from aged garlic on liver cell injury. A chronic ethanol-fed ALD in vivo model (the NIAAA model) was used to test the protective functions of SAMC. It was observed that SAMC (300 mg/kg, by gavage method) effectively ameliorated ALD-induced body weight reduction, steatosis, insulin resistance, and inflammation without affecting the health status of the control mice, as demonstrated by histological, biochemical, and molecular biology assays. By using biophysical assays and molecular docking, we demonstrated that SAMC directly targeted insulin receptor (INSR) protein on the cell membrane and then restored downstream IRS-1/AKT/GSK3β signaling. Liver-specific knock-down in mice and siRNA-mediated knock-down in AML-12 cells of Insr significantly impaired SAMC (250 μmol/L in cells)-mediated protection. Restoration of the IRS-1/AKT signaling partly recovered hepatic injury and further contributed to SAMC's beneficial effects. Continuous administration of AKT agonist and recombinant IGF-1 in combination with SAMC showed hepato-protection in the mice model. Long-term (90-day) administration of SAMC had no obvious adverse effect on healthy mice. We conclude that SAMC is an effective and safe hepato-protective complimentary agent against ALD partly through the direct binding of INSR and partial regulation of the IRS-1/AKT/GSK3β pathway.
Collapse
Key Words
- ADIPOQ, adiponectin
- AKT
- ALD, alcoholic liver disease
- ALDH2, aldehyde dehydrogenase 2
- ALT, alanine aminotransferase
- AMPK, adenosine 5′-monophosphate (AMP)-activated protein kinase
- AST, aspartate aminotransferase
- ATGL, adipose triglyceride lipase
- Alcoholic liver disease
- CPT1, carnitine palmitoyltransferase I
- CYP2E1, cytochrome P450 2E1
- FDA, U.S. Food and Drug Administration
- FFA, free fatty acids
- GRB14, growth factor receptor-bound protein 14
- GSK3β
- GSK3β, glycogen synthase kinase 3 beta
- GTT, glucose tolerance test
- HSL, hormone sensitive lipase
- IGF-1, insulin-like growth factors-1
- IL, interleukin
- INSR, insulin receptor
- IRS, insulin receptor substrate
- IRS-1
- IRTK, insulin receptor tyrosine kinase
- Insulin receptor
- Insulin resistance
- LDLR, low-density lipoprotein receptor
- LRP6, low-density lipoprotein receptor related protein 6
- MTT, 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyl-tetrazolium bromide
- NAC, N-acetyl-cysteine
- NAFLD, non-alcoholic fatty liver disease
- NAS, NAFLD activity score
- NF-κB, nuclear factor kappa B
- NIAAA, National Institute on Alcohol Abuse and Alcoholism
- NRF2, nuclear factor erythroid 2-related factor 2
- ORF, open reading frame
- PA, palmitate acid
- PPARα, peroxisome proliferator-activated receptor alpha
- RER, respiratory exchange ratio
- S-Allylmercaptocysteine
- SAMC, S-allylmercaptocysteine
- SPR, surface plasmon resonance
- SREBP-1c, sterol regulatory element-binding protein 1c
- Safety
- TC, total cholesterol
- TCF/LEF, T-cell factor/lymphoid enhancer factor
- TG, triglyceride
- TNF, tumor necrosis factor
- TSA, thermal shift assay
- WAT, white adipose tissues
- WT, wild-type
Collapse
|
11
|
Pei J, Xiao Z, Guo Z, Pei Y, Wei S, Wu H, Wang D. Sustained Stimulation of β 2AR Inhibits Insulin Signaling in H9C2 Cardiomyoblast Cells Through the PKA-Dependent Signaling Pathway. Diabetes Metab Syndr Obes 2020; 13:3887-3898. [PMID: 33116735 PMCID: PMC7585860 DOI: 10.2147/dmso.s268028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/05/2020] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION This study aimed to investigate the role of β2 adrenergic receptor (β2AR) in insulin signaling transduction in H9C2 cardiomyoblast cells to understand the formation of the β2AR-insulin receptor (IR) protein complex and its role in insulin-induced Glut4 expression. METHODS H9C2 cells were treated with various protein inhibitors (CGP, β1AR inhibitor CGP20712; ICI, β2AR inhibitor ICI 118,551; PKI, PKA inhibitor myristoylated PKI; PD 0325901, MEK inhibitor; SP600125, JNK inhibitor) with or without insulin or isoproterenol (ISO) before RNA-sequencing (RNA-Seq) and quantitative-PCR (Q-PCR). Yeast two-hybrid, co-immunoprecipitation and His-tag pull-down assay were carried out to investigate the formation of the β2AR-IR protein complex. The intracellular concentrations of cAMP in H9C2 cells were tested by high performance liquid chromatography (HPLC) and the phosphorylation of JNK was tested by Western blot. RESULTS Gene Ontology (GO) analysis revealed that the most significantly enriched processes in the domain of molecular function (MF) were catalytic activity and binding, whereas in the domain of biological processes (BP) were metabolic process and cellular process. Furthermore, the enriched processes in the domain of cellular components (CC) were cell and cell parts. The Kyoto encyclopedia of genes and genomes (KEGG) pathway analysis showed that the most significant pathways that have been altered included the PI3K-Akt and MAPK signaling pathways. Q-PCR, which was performed to verify the gene expression levels exhibited consistent results. In evaluating the signaling pathways, the sustained stimulation of β2AR by ISO inhibited insulin signalling, and the effect was primarily through the cAMP-PKA-JNK pathway and MEK/JNK signaling pathway. Yeast two-hybrid, co-immunoprecipitation and His-tag pull-down assay revealed that β2AR, IR, insulin receptor substrate 1 (IRS1), Grb2-associated binding protein 1 (GAB1) and Grb2 existed in the same protein complex. CONCLUSION The sustained stimulation of β2AR might inhibit insulin signaling transduction through the cAMP-PKA-JNK and MEK/JNK pathways in H9C2 cells.
Collapse
Affiliation(s)
- Jinli Pei
- Key Laboratory of Ministry of Education for Tropical Bioresources, Hainan University, Haikou, Hainan570228, People's Republic of China
- Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan570228, People's Republic of China
| | - Zhengpan Xiao
- Key Laboratory of Ministry of Education for Tropical Bioresources, Hainan University, Haikou, Hainan570228, People's Republic of China
- Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan570228, People's Republic of China
| | - Ziyi Guo
- Key Laboratory of Ministry of Education for Tropical Bioresources, Hainan University, Haikou, Hainan570228, People's Republic of China
- Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan570228, People's Republic of China
| | - Yechun Pei
- Key Laboratory of Ministry of Education for Tropical Bioresources, Hainan University, Haikou, Hainan570228, People's Republic of China
- Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan570228, People's Republic of China
| | - Shuangshuang Wei
- Key Laboratory of Ministry of Education for Tropical Bioresources, Hainan University, Haikou, Hainan570228, People's Republic of China
- Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan570228, People's Republic of China
| | - Hao Wu
- Key Laboratory of Ministry of Education for Tropical Bioresources, Hainan University, Haikou, Hainan570228, People's Republic of China
- Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan570228, People's Republic of China
| | - Dayong Wang
- Key Laboratory of Ministry of Education for Tropical Bioresources, Hainan University, Haikou, Hainan570228, People's Republic of China
- Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan570228, People's Republic of China
| |
Collapse
|
12
|
Ding X, Iyer R, Novotny C, Metzger D, Zhou HH, Smith GI, Yoshino M, Yoshino J, Klein S, Swaminath G, Talukdar S, Zhou Y. Inhibition of Grb14, a negative modulator of insulin signaling, improves glucose homeostasis without causing cardiac dysfunction. Sci Rep 2020; 10:3417. [PMID: 32099031 PMCID: PMC7042267 DOI: 10.1038/s41598-020-60290-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 02/09/2020] [Indexed: 01/25/2023] Open
Abstract
Insulin resistance increases patients' risk of developing type 2 diabetes (T2D), non-alcoholic steatohepatitis (NASH) and a host of other comorbidities including cardiovascular disease and cancer. At the molecular level, insulin exerts its function through the insulin receptor (IR), a transmembrane receptor tyrosine kinase. Data from human genetic studies have shown that Grb14 functions as a negative modulator of IR activity, and the germline Grb14-knockout (KO) mice have improved insulin signaling in liver and skeletal muscle. Here, we show that Grb14 knockdown in liver, white adipose tissues, and heart with an AAV-shRNA (Grb14-shRNA) improves glucose homeostasis in diet-induced obese (DIO) mice. A previous report has shown that germline deletion of Grb14 in mice results in cardiac hypertrophy and impaired systolic function, which could severely limit the therapeutic potential of targeting Grb14. In this report, we demonstrate that there are no significant changes in cardiac function as measured by echocardiography in the Grb14-knockdown mice fed a high-fat diet for a period of four months. While additional studies are needed to further confirm the efficacy and to de-risk potential negative cardiac effects in preclinical models, our data support the therapeutic strategy of inhibiting Grb14 to treat diabetes and related conditions.
Collapse
Affiliation(s)
| | | | | | | | | | - Gordon I Smith
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Mihoko Yoshino
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jun Yoshino
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Samuel Klein
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | |
Collapse
|
13
|
Taira J, Yoshida K, Takemoto M, Hanada K, Sakamoto H. Dephosphorylation of clustered phosphoserine residues in human Grb14 by protein phosphatase 1 and its effect on insulin receptor complex formation. J Pept Sci 2019; 25:e3207. [PMID: 31347216 DOI: 10.1002/psc.3207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/20/2019] [Accepted: 07/06/2019] [Indexed: 11/10/2022]
Abstract
The physical interaction of the human growth factor receptor-bound protein 14 (hGrb14) and the insulin receptor (IR) represses insulin signaling. With respect to the recruiting mechanism of hGrb14 to IR respond to insulin stimulus, our previous reports have suggested that phosphorylation of Ser358 , Ser362 , and Ser366 in hGrb14 by glycogen synthase kinase-3 repressed hGrb14-IR complex formation. In this study, we investigated phosphatase-mediated dephosphorylation of the hGrb14 phosphoserine residues. An in vitro phosphatase assay with hGrb14-derived synthetic phosphopeptides suggested that protein phosphatase 1 (PP1) is involved in the dephosphorylation of Ser358 and Ser362 . Furthermore, coimmunoprecipitation experiments suggested that insulin-induced hGrb14-IR complex formation was repressed by the substitution of Ser358 or Ser362 with glutamic acid. These findings suggested that phosphate groups on Ser358 and Ser362 in hGrb14 are dephosphorylated by PP1, and the dephosphorylation facilitates hGrb14-IR complex formation.
Collapse
Affiliation(s)
- Junichi Taira
- Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, Iizuka, Japan
| | - Keisuke Yoshida
- Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, Iizuka, Japan
| | - Misaki Takemoto
- Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, Iizuka, Japan
| | - Kousuke Hanada
- Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, Iizuka, Japan
| | - Hiroshi Sakamoto
- Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, Iizuka, Japan
| |
Collapse
|
14
|
Chu PY, Tai YL, Shen TL. Grb7, a Critical Mediator of EGFR/ErbB Signaling, in Cancer Development and as a Potential Therapeutic Target. Cells 2019; 8:cells8050435. [PMID: 31083325 PMCID: PMC6562560 DOI: 10.3390/cells8050435] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 05/02/2019] [Accepted: 05/09/2019] [Indexed: 12/20/2022] Open
Abstract
The partner of activated epidermal growth factor receptor (EGFR), growth factor receptor bound protein-7 (Grb7), a functionally multidomain adaptor protein, has been demonstrated to be a pivotal regulator for varied physiological and pathological processes by interacting with phospho-tyrosine-related signaling molecules to affect the transmission through a number of signaling pathways. In particular, critical roles of Grb7 in erythroblastic leukemia viral oncogene homolog (ERBB) family-mediated cancer development and malignancy have been intensively evaluated. The overexpression of Grb7 or the coamplification/cooverexpression of Grb7 and members of the ERBB family play essential roles in advanced human cancers and are associated with decreased survival and recurrence of cancers, emphasizing Grb7's value as a prognostic marker and a therapeutic target. Peptide inhibitors of Grb7 are being tested in preclinical trials for their possible therapeutic effects. Here, we review the molecular, functional, and clinical aspects of Grb7 in ERBB family-mediated cancer development and malignancy with the aim to reveal alternative and effective therapeutic strategies.
Collapse
Affiliation(s)
- Pei-Yu Chu
- Department of Plant Pathology and Microbiology, National Taiwan University, Taipei 10617, Taiwan.
| | - Yu-Ling Tai
- Department of Plant Pathology and Microbiology, National Taiwan University, Taipei 10617, Taiwan.
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Tang-Long Shen
- Department of Plant Pathology and Microbiology, National Taiwan University, Taipei 10617, Taiwan.
- Center for Biotechnology, National Taiwan University, Taipei 10617, Taiwan.
| |
Collapse
|
15
|
Rabiee N, Bagherzadeh M, Rabiee M. A Perspective to the Correlation Between Brain Insulin Resistance and Alzheimer: Medicinal Chemistry Approach. Curr Diabetes Rev 2019; 15:255-258. [PMID: 30381082 DOI: 10.2174/1573399814666181031154817] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/17/2018] [Accepted: 10/29/2018] [Indexed: 12/11/2022]
Abstract
Substantial terms have been recognized on the associated risk elements, comorbidities as well as, putative pathophysiological processes of Alzheimer disease and related dementias (ADRDs) as well as, type 2 diabetes mellitus (T2DM), a few from greatest important disease from the moments. Very much is considered regarding the biology and chemistry of each predicament, nevertheless T2DM and ADRDs are an actually similar pattern developing from the similar origins of maturing or synergistic conditions connected by aggressive patho-corporeal terms and continues to be ambiguous. In this depth-critique article, we aimed to investigate all possibilities and represented a novel and applicable approach from the Medicinal Chemistry concepts.
Collapse
Affiliation(s)
- Navid Rabiee
- Department of Chemistry, Sharif University of Technology, Tehran, Iran
| | | | - Mohammad Rabiee
- Biomaterial Group, Biomedical Engineering Department, Amirkabir University of Technology, Tehran, Iran
| |
Collapse
|
16
|
Liau NPD, Laktyushin A, Lucet IS, Murphy JM, Yao S, Whitlock E, Callaghan K, Nicola NA, Kershaw NJ, Babon JJ. The molecular basis of JAK/STAT inhibition by SOCS1. Nat Commun 2018. [PMID: 29674694 DOI: 10.1038/s41467‐018‐04013‐1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The SOCS family of proteins are negative-feedback inhibitors of signalling induced by cytokines that act via the JAK/STAT pathway. SOCS proteins can act as ubiquitin ligases by recruiting Cullin5 to ubiquitinate signalling components; however, SOCS1, the most potent member of the family, can also inhibit JAK directly. Here we determine the structural basis of both these modes of inhibition. Due to alterations within the SOCS box domain, SOCS1 has a compromised ability to recruit Cullin5; however, it is a direct, potent and selective inhibitor of JAK catalytic activity. The kinase inhibitory region of SOCS1 targets the substrate binding groove of JAK with high specificity and thereby blocks any subsequent phosphorylation. SOCS1 is a potent inhibitor of the interferon gamma (IFNγ) pathway, however, it does not bind the IFNγ receptor, making its mode-of-action distinct from SOCS3. These findings reveal the mechanism used by SOCS1 to inhibit signalling by inflammatory cytokines.
Collapse
Affiliation(s)
- Nicholas P D Liau
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC, 3052, Australia.,The University of Melbourne, Royal Parade, Parkville, VIC, 3050, Australia
| | - Artem Laktyushin
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC, 3052, Australia.,The University of Melbourne, Royal Parade, Parkville, VIC, 3050, Australia
| | - Isabelle S Lucet
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC, 3052, Australia.,The University of Melbourne, Royal Parade, Parkville, VIC, 3050, Australia
| | - James M Murphy
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC, 3052, Australia.,The University of Melbourne, Royal Parade, Parkville, VIC, 3050, Australia
| | - Shenggen Yao
- The University of Melbourne, Royal Parade, Parkville, VIC, 3050, Australia
| | - Eden Whitlock
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC, 3052, Australia.,The University of Melbourne, Royal Parade, Parkville, VIC, 3050, Australia
| | - Kimberley Callaghan
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC, 3052, Australia.,The University of Melbourne, Royal Parade, Parkville, VIC, 3050, Australia
| | - Nicos A Nicola
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC, 3052, Australia.,The University of Melbourne, Royal Parade, Parkville, VIC, 3050, Australia
| | - Nadia J Kershaw
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC, 3052, Australia. .,The University of Melbourne, Royal Parade, Parkville, VIC, 3050, Australia.
| | - Jeffrey J Babon
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC, 3052, Australia. .,The University of Melbourne, Royal Parade, Parkville, VIC, 3050, Australia.
| |
Collapse
|
17
|
Liau NPD, Laktyushin A, Lucet IS, Murphy JM, Yao S, Whitlock E, Callaghan K, Nicola NA, Kershaw NJ, Babon JJ. The molecular basis of JAK/STAT inhibition by SOCS1. Nat Commun 2018; 9:1558. [PMID: 29674694 PMCID: PMC5908791 DOI: 10.1038/s41467-018-04013-1] [Citation(s) in RCA: 304] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 03/27/2018] [Indexed: 12/22/2022] Open
Abstract
The SOCS family of proteins are negative-feedback inhibitors of signalling induced by cytokines that act via the JAK/STAT pathway. SOCS proteins can act as ubiquitin ligases by recruiting Cullin5 to ubiquitinate signalling components; however, SOCS1, the most potent member of the family, can also inhibit JAK directly. Here we determine the structural basis of both these modes of inhibition. Due to alterations within the SOCS box domain, SOCS1 has a compromised ability to recruit Cullin5; however, it is a direct, potent and selective inhibitor of JAK catalytic activity. The kinase inhibitory region of SOCS1 targets the substrate binding groove of JAK with high specificity and thereby blocks any subsequent phosphorylation. SOCS1 is a potent inhibitor of the interferon gamma (IFNγ) pathway, however, it does not bind the IFNγ receptor, making its mode-of-action distinct from SOCS3. These findings reveal the mechanism used by SOCS1 to inhibit signalling by inflammatory cytokines. Cytokines are key molecules in controlling haematopoiesis that signal via the JAK/STAT pathway. Here the authors present the structures of SOCS1 bound to its JAK1 target as well as in complex with elonginB and elonginC, providing a molecular explanation for the potent JAK- inhibitory activity of SOCS1.
Collapse
Affiliation(s)
- Nicholas P D Liau
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC, 3052, Australia.,The University of Melbourne, Royal Parade, Parkville, VIC, 3050, Australia
| | - Artem Laktyushin
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC, 3052, Australia.,The University of Melbourne, Royal Parade, Parkville, VIC, 3050, Australia
| | - Isabelle S Lucet
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC, 3052, Australia.,The University of Melbourne, Royal Parade, Parkville, VIC, 3050, Australia
| | - James M Murphy
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC, 3052, Australia.,The University of Melbourne, Royal Parade, Parkville, VIC, 3050, Australia
| | - Shenggen Yao
- The University of Melbourne, Royal Parade, Parkville, VIC, 3050, Australia
| | - Eden Whitlock
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC, 3052, Australia.,The University of Melbourne, Royal Parade, Parkville, VIC, 3050, Australia
| | - Kimberley Callaghan
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC, 3052, Australia.,The University of Melbourne, Royal Parade, Parkville, VIC, 3050, Australia
| | - Nicos A Nicola
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC, 3052, Australia.,The University of Melbourne, Royal Parade, Parkville, VIC, 3050, Australia
| | - Nadia J Kershaw
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC, 3052, Australia. .,The University of Melbourne, Royal Parade, Parkville, VIC, 3050, Australia.
| | - Jeffrey J Babon
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC, 3052, Australia. .,The University of Melbourne, Royal Parade, Parkville, VIC, 3050, Australia.
| |
Collapse
|
18
|
Holt LJ, Brandon AE, Small L, Suryana E, Preston E, Wilks D, Mokbel N, Coles CA, White JD, Turner N, Daly RJ, Cooney GJ. Ablation of Grb10 Specifically in Muscle Impacts Muscle Size and Glucose Metabolism in Mice. Endocrinology 2018; 159:1339-1351. [PMID: 29370381 DOI: 10.1210/en.2017-00851] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 01/17/2018] [Indexed: 12/14/2022]
Abstract
Grb10 is an adaptor-type signaling protein most highly expressed in tissues involved in insulin action and glucose metabolism, such as muscle, pancreas, and adipose. Germline deletion of Grb10 in mice creates a phenotype with larger muscles and improved glucose homeostasis. However, it has not been determined whether Grb10 ablation specifically in muscle is sufficient to induce hypermuscularity or affect whole body glucose metabolism. In this study we generated muscle-specific Grb10-deficient mice (Grb10-mKO) by crossing Grb10flox/flox mice with mice expressing Cre recombinase under control of the human α-skeletal actin promoter. One-year-old Grb10-mKO mice had enlarged muscles, with greater cross-sectional area of fibers compared with wild-type (WT) mice. This degree of hypermuscularity did not affect whole body glucose homeostasis under basal conditions. However, hyperinsulinemic/euglycemic clamp studies revealed that Grb10-mKO mice had greater glucose uptake into muscles compared with WT mice. Insulin signaling was increased at the level of phospho-Akt in muscle of Grb10-mKO mice compared with WT mice, consistent with a role of Grb10 as a modulator of proximal insulin receptor signaling. We conclude that ablation of Grb10 in muscle is sufficient to affect muscle size and metabolism, supporting an important role for this protein in growth and metabolic pathways.
Collapse
Affiliation(s)
- Lowenna J Holt
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Amanda E Brandon
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- School of Medical Sciences, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Lewin Small
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Eurwin Suryana
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Elaine Preston
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Donna Wilks
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Nancy Mokbel
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Chantal A Coles
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Jason D White
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Veterinary Biosciences, Faculty of Veterinary and Agricultural Science, University of Melbourne, Parkville, Victoria, Australia
| | - Nigel Turner
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Department of Pharmacology, University of New South Wales, Sydney, New South Wales, Australia
| | - Roger J Daly
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Gregory J Cooney
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- School of Medical Sciences, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
19
|
A systematic analysis highlights multiple long non-coding RNAs associated with cardiometabolic disorders. J Hum Genet 2018; 63:431-446. [PMID: 29382920 DOI: 10.1038/s10038-017-0403-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 12/19/2022]
Abstract
Genome-wide association studies (GWAS) have identified many susceptibility loci for cardiometabolic disorders. Most of the associated variants reside in non-coding regions of the genome including long non-coding RNAs (lncRNAs), which are thought to play critical roles in diverse biological processes. Here, we leveraged data from the available GWAS meta-analyses on lipid and obesity-related traits, blood pressure, type 2 diabetes, and coronary artery disease and identified 179 associated single-nucleotide polymorphisms (SNPs) in 102 lncRNAs (p-value < 2.3 × 10-7). Of these, 55 SNPs, either the lead SNP or in strong linkage disequilibrium with the lead SNP in the related loci, were selected for further investigations. Our in silico predictions and functional annotations of the SNPs as well as expression and DNA methylation analysis of their lncRNAs demonstrated several lncRNAs that fulfilled predefined criteria for being potential functional targets. In particular, we found evidence suggesting that LOC157273 (at 8p23.1) is involved in regulating serum lipid-cholesterol. Our results showed that rs4841132 in the second exon and cg17371580 in the promoter region of LOC157273 are associated with lipids; the lncRNA is expressed in liver and associates with the expression of its nearby coding gene, PPP1R3B. Collectively, we highlight a number of loci associated with cardiometabolic disorders for which the association may act through lncRNAs.
Collapse
|
20
|
Activation of oncogenic tyrosine kinase signaling promotes insulin receptor-mediated cone photoreceptor survival. Oncotarget 2018; 7:46924-46942. [PMID: 27391439 PMCID: PMC5216914 DOI: 10.18632/oncotarget.10447] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 06/26/2016] [Indexed: 01/18/2023] Open
Abstract
In humans, daylight vision is primarily mediated by cone photoreceptors. These cells die in age-related retinal degenerations. Prolonging the life of cones for even one decade would have an enormous beneficial effect on usable vision in an aging population. Photoreceptors are postmitotic, but shed 10% of their outer segments daily, and must synthesize the membrane and protein equivalent of a proliferating cell each day. Although activation of oncogenic tyrosine kinase and inhibition of tyrosine phosphatase signaling is known to be essential for tumor progression, the cellular regulation of this signaling in postmitotic photoreceptor cells has not been studied. In the present study, we report that a novel G-protein coupled receptor–mediated insulin receptor (IR) signaling pathway is regulated by non-receptor tyrosine kinase Src through the inhibition of protein tyrosine phosphatase IB (PTP1B). We demonstrated the functional significance of this pathway through conditional deletion of IR and PTP1B in cones, in addition to delaying the death of cones in a mouse model of cone degeneration by activating the Src. This is the first study demonstrating the molecular mechanism of a novel signaling pathway in photoreceptor cells, which provides a window of opportunity to save the dying cones in retinal degenerative diseases.
Collapse
|
21
|
Haeusler RA, McGraw TE, Accili D. Biochemical and cellular properties of insulin receptor signalling. Nat Rev Mol Cell Biol 2018; 19:31-44. [PMID: 28974775 PMCID: PMC5894887 DOI: 10.1038/nrm.2017.89] [Citation(s) in RCA: 472] [Impact Index Per Article: 67.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The mechanism of insulin action is a central theme in biology and medicine. In addition to the rather rare condition of insulin deficiency caused by autoimmune destruction of pancreatic β-cells, genetic and acquired abnormalities of insulin action underlie the far more common conditions of type 2 diabetes, obesity and insulin resistance. The latter predisposes to diseases ranging from hypertension to Alzheimer disease and cancer. Hence, understanding the biochemical and cellular properties of insulin receptor signalling is arguably a priority in biomedical research. In the past decade, major progress has led to the delineation of mechanisms of glucose transport, lipid synthesis, storage and mobilization. In addition to direct effects of insulin on signalling kinases and metabolic enzymes, the discovery of mechanisms of insulin-regulated gene transcription has led to a reassessment of the general principles of insulin action. These advances will accelerate the discovery of new treatment modalities for diabetes.
Collapse
Affiliation(s)
- Rebecca A Haeusler
- Columbia University College of Physicians and Surgeons, Department of Pathology and Cell Biology, New York, New York 10032, USA
| | - Timothy E McGraw
- Weill Cornell Medicine, Departments of Biochemistry and Cardiothoracic Surgery, New York, New York 10065, USA
| | - Domenico Accili
- Columbia University College of Physicians & Surgeons, Department of Medicine, New York, New York 10032, USA
| |
Collapse
|
22
|
Villalobo A, Ishida H, Vogel HJ, Berchtold MW. Calmodulin as a protein linker and a regulator of adaptor/scaffold proteins. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1865:507-521. [PMID: 29247668 DOI: 10.1016/j.bbamcr.2017.12.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/07/2017] [Accepted: 12/08/2017] [Indexed: 01/29/2023]
Abstract
Calmodulin (CaM) is a universal regulator for a huge number of proteins in all eukaryotic cells. Best known is its function as a calcium-dependent modulator of the activity of enzymes, such as protein kinases and phosphatases, as well as other signaling proteins including membrane receptors, channels and structural proteins. However, less well known is the fact that CaM can also function as a Ca2+-dependent adaptor protein, either by bridging between different domains of the same protein or by linking two identical or different target proteins together. These activities are possible due to the fact that CaM contains two independently-folded Ca2+ binding lobes that are able to interact differentially and to some degree separately with targets proteins. In addition, CaM can interact with and regulates several proteins that function exclusively as adaptors. This review provides an overview over our present knowledge concerning the structural and functional aspects of the role of CaM as an adaptor protein and as a regulator of known adaptor/scaffold proteins.
Collapse
Affiliation(s)
- Antonio Villalobo
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Arturo Duperier 4, E-28029 Madrid, Spain.
| | - Hiroaki Ishida
- Department of Biological Sciences, University of Calgary, 2500 University Dr. N.W., Calgary, Alberta T2N 1N4, Canada
| | - Hans J Vogel
- Department of Biological Sciences, University of Calgary, 2500 University Dr. N.W., Calgary, Alberta T2N 1N4, Canada.
| | - Martin W Berchtold
- Department of Biology, University of Copenhagen, 13 Universitetsparken, DK-2100 Copenhagen Ø, Denmark.
| |
Collapse
|
23
|
Naudin C, Chevalier C, Roche S. The role of small adaptor proteins in the control of oncogenic signalingr driven by tyrosine kinases in human cancer. Oncotarget 2017; 7:11033-55. [PMID: 26788993 PMCID: PMC4905456 DOI: 10.18632/oncotarget.6929] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 01/01/2016] [Indexed: 12/15/2022] Open
Abstract
Protein phosphorylation on tyrosine (Tyr) residues has evolved as an important mechanism to coordinate cell communication in multicellular organisms. The importance of this process has been revealed by the discovery of the prominent oncogenic properties of tyrosine kinases (TK) upon deregulation of their physiological activities, often due to protein overexpression and/or somatic mutation. Recent reports suggest that TK oncogenic signaling is also under the control of small adaptor proteins. These cytosolic proteins lack intrinsic catalytic activity and signal by linking two functional members of a catalytic pathway. While most adaptors display positive regulatory functions, a small group of this family exerts negative regulatory functions by targeting several components of the TK signaling cascade. Here, we review how these less studied adaptor proteins negatively control TK activities and how their loss of function induces abnormal TK signaling, promoting tumor formation. We also discuss the therapeutic consequences of this novel regulatory mechanism in human oncology.
Collapse
Affiliation(s)
- Cécile Naudin
- CNRS UMR5237, University Montpellier, CRBM, Montpellier, France.,Present address: INSERM U1016, CNRS UMR8104, Institut Cochin, Paris, France
| | - Clément Chevalier
- CNRS UMR5237, University Montpellier, CRBM, Montpellier, France.,Present address: SFR Biosit (UMS CNRS 3480/US INSERM 018), MRic Photonics Platform, University Rennes, Rennes, France
| | - Serge Roche
- CNRS UMR5237, University Montpellier, CRBM, Montpellier, France.,Equipe Labellisée LIGUE 2014, Ligue Contre le Cancer, Paris, France
| |
Collapse
|
24
|
Gondoin A, Hampe C, Eudes R, Fayolle C, Pierre-Eugène C, Miteva M, Villoutreix BO, Charnay-Pouget F, Aitken DJ, Issad T, Burnol AF. Identification of insulin-sensitizing molecules acting by disrupting the interaction between the Insulin Receptor and Grb14. Sci Rep 2017; 7:16901. [PMID: 29203791 PMCID: PMC5715071 DOI: 10.1038/s41598-017-17122-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 11/22/2017] [Indexed: 01/07/2023] Open
Abstract
Metabolic diseases are characterized by a decreased action of insulin. During the course of the disease, usual treatments frequently fail and patients are finally submitted to insulinotherapy. There is thus a need for innovative therapeutic strategies to improve insulin action. Growth factor receptor-bound protein 14 (Grb14) is a molecular adapter that specifically binds to the activated insulin receptor (IR) and inhibits its tyrosine kinase activity. Molecules disrupting Grb14-IR binding are therefore potential insulin-sensitizing agents. We used Structure-Based Virtual Ligand Screening to generate a list of 1000 molecules predicted to hinder Grb14-IR binding. Using an acellular bioluminescence resonance energy transfer (BRET) assay, we identified, out of these 1000 molecules, 3 compounds that inhibited Grb14-IR interaction. Their inhibitory effect on insulin-induced Grb14-IR interaction was confirmed in co-immunoprecipitation experiments. The more efficient molecule (C8) was further characterized. C8 increased downstream Ras-Raf and PI3-kinase insulin signaling, as shown by BRET experiments in living cells. Moreover, C8 regulated the expression of insulin target genes in mouse primary hepatocytes. These results indicate that C8, by reducing Grb14-IR interaction, increases insulin signalling. The use of C8 as a lead compound should allow for the development of new molecules of potential therapeutic interest for the treatment of diabetes.
Collapse
Affiliation(s)
- Anaïs Gondoin
- Institut Cochin, Université Paris Descartes, CNRS (UMR8104), Paris, France.,INSERM, U1016, Paris, France
| | - Cornelia Hampe
- Institut Cochin, Université Paris Descartes, CNRS (UMR8104), Paris, France.,INSERM, U1016, Paris, France
| | - Richard Eudes
- Université Paris Diderot, Sorbonne-Paris-Cité, Inserm UMR-S 973, Molécules Thérapeutiques in silico, Paris, France
| | - Cyril Fayolle
- Institut Cochin, Université Paris Descartes, CNRS (UMR8104), Paris, France.,INSERM, U1016, Paris, France
| | - Cécile Pierre-Eugène
- Institut Cochin, Université Paris Descartes, CNRS (UMR8104), Paris, France.,INSERM, U1016, Paris, France
| | - Maria Miteva
- Université Paris Diderot, Sorbonne-Paris-Cité, Inserm UMR-S 973, Molécules Thérapeutiques in silico, Paris, France
| | - Bruno O Villoutreix
- Université Paris Diderot, Sorbonne-Paris-Cité, Inserm UMR-S 973, Molécules Thérapeutiques in silico, Paris, France
| | - Florence Charnay-Pouget
- CP3A Organic Synthesis Group, ICMMO, UMR 8182, CNRS, Université Paris Sud, Université Paris Saclay, Orsay, France
| | - David J Aitken
- CP3A Organic Synthesis Group, ICMMO, UMR 8182, CNRS, Université Paris Sud, Université Paris Saclay, Orsay, France
| | - Tarik Issad
- Institut Cochin, Université Paris Descartes, CNRS (UMR8104), Paris, France. .,INSERM, U1016, Paris, France.
| | - Anne-Françoise Burnol
- Institut Cochin, Université Paris Descartes, CNRS (UMR8104), Paris, France. .,INSERM, U1016, Paris, France.
| |
Collapse
|
25
|
Abstract
Insulin resistance and the metabolic syndrome are complex metabolic traits and key risk factors for the development of cardiovascular disease. They result from the interplay of environmental and genetic factors but the full extent of the genetic background to these conditions remains incomplete. Large-scale genome-wide association studies have helped advance the identification of common genetic variation associated with insulin resistance and the metabolic syndrome, and more recently, exome sequencing has allowed the identification of rare variants associated with the pathogenesis of these conditions. Many variants associated with insulin resistance are directly involved in glucose metabolism; however, functional studies are required to assess the contribution of other variants to the development of insulin resistance. Many genetic variants involved in the pathogenesis of the metabolic syndrome are associated with lipid metabolism.
Collapse
Affiliation(s)
- Audrey E Brown
- Institute of Cellular Medicine, William Leech Building, Medical School, Newcastle University, Newcastle, NE2 4HH, UK
| | - Mark Walker
- Institute of Cellular Medicine, William Leech Building, Medical School, Newcastle University, Newcastle, NE2 4HH, UK.
| |
Collapse
|
26
|
Ye L, Maji S, Sanghera N, Gopalasingam P, Gorbunov E, Tarasov S, Epstein O, Klein-Seetharaman J. Structure and dynamics of the insulin receptor: implications for receptor activation and drug discovery. Drug Discov Today 2017; 22:1092-1102. [PMID: 28476537 DOI: 10.1016/j.drudis.2017.04.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 04/16/2017] [Accepted: 04/19/2017] [Indexed: 01/05/2023]
Abstract
Recently, major progress has been made in uncovering the mechanisms of how insulin engages its receptor and modulates downstream signal transduction. Here, we present in detail the current structural knowledge surrounding the individual components of the complex, binding sites, and dynamics during the activation process. A novel kinase triggering mechanism, the 'bow-arrow model', is proposed based on current knowledge and computational simulations of this system, in which insulin, after its initial interaction with binding site 1, engages with site 2 between the fibronectin type III (FnIII)-1 and -2 domains, which changes the conformation of FnIII-3 and eventually translates into structural changes across the membrane. This model provides a new perspective on the process of insulin binding to its receptor and, thus, could lead to future novel drug discovery efforts.
Collapse
Affiliation(s)
- Libin Ye
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Suvrajit Maji
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Narinder Sanghera
- Division of Metabolic and Vascular Health & Systems, Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Piraveen Gopalasingam
- Division of Metabolic and Vascular Health & Systems, Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Evgeniy Gorbunov
- OOO 'NPF 'MATERIA MEDICA HOLDING', 47-1, Trifonovskaya St, Moscow 129272, Russian Federation
| | - Sergey Tarasov
- OOO 'NPF 'MATERIA MEDICA HOLDING', 47-1, Trifonovskaya St, Moscow 129272, Russian Federation
| | - Oleg Epstein
- The Institute of General Pathology and Pathophysiology, 8, Baltiyskaya St, 125315 Moscow, Russian Federation
| | - Judith Klein-Seetharaman
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA; Division of Metabolic and Vascular Health & Systems, Medical School, University of Warwick, Coventry CV4 7AL, UK.
| |
Collapse
|
27
|
García-Palmero I, Pompas-Veganzones N, Villalobo E, Gioria S, Haiech J, Villalobo A. The adaptors Grb10 and Grb14 are calmodulin-binding proteins. FEBS Lett 2017; 591:1176-1186. [PMID: 28295264 DOI: 10.1002/1873-3468.12623] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/06/2017] [Indexed: 01/24/2023]
Abstract
We identified the Grb7 family members, Grb10 and Grb14, as Ca2+ -dependent CaM-binding proteins using Ca2+ -dependent CaM-affinity chromatography as we previously did with Grb7. The potential CaM-binding sites were identified and experimentally tested using fluorescent-labeled peptides corresponding to these sites. The apparent affinity constant of these peptides for CaM, and the minimum number of calcium ions bound to CaM that are required for effective binding to these peptides were also determined. We prepared deletion mutants of the three adaptor proteins lacking the identified sites and determined that they lost or strongly diminished their CaM-binding capacity following the sequence Grb7 > > Grb14 > Grb10. More than one CaM-binding site and/or accessory CaM-binding sites appear to exist in Grb10 and Grb14, as compared to a single one present in Grb7.
Collapse
Affiliation(s)
- Irene García-Palmero
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Spain
| | - Noemí Pompas-Veganzones
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Spain
| | - Eduardo Villalobo
- Departamento de Microbiología, Facultad de Biología, Universidad de Sevilla, Spain
| | - Sophie Gioria
- Plate-forme de Chimie Biologique Intégrative de Strasbourg (PCBIS), UMS 3286 CNRS-Université de Strasbourg, France
| | - Jacques Haiech
- Laboratoire d'Excellence Medalis, Université de Strasbourg, CNRS, LIT UMR 7200, France
| | - Antonio Villalobo
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Spain
| |
Collapse
|
28
|
Chatchaiphan S, Srisapoome P, Kim JH, Devlin RH, Na-Nakorn U. De Novo Transcriptome Characterization and Growth-Related Gene Expression Profiling of Diploid and Triploid Bighead Catfish (Clarias macrocephalus Günther, 1864). MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2017; 19:36-48. [PMID: 28181037 DOI: 10.1007/s10126-017-9730-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 12/31/2016] [Indexed: 06/06/2023]
Abstract
To enhance understanding of triploid gene expression, the transcriptome information from bighead catfish (Clarias macrocephalus Günther, 1864) was studied using the paired-end Illumina HiSeq™ 2000 sequencing platform. In total, 68,227,832 raw reads were generated from liver tissues and 53,149 unigenes were assembled, with an average length of 765 bp and N50 length of 1283 bp. Of these unigenes, 33,428 (62.89%) could be annotated according to their homology with matches in the NCBI non-redundant (Nr), NCBI nucleotide (Nt), Swiss-Prot, Clusters of Orthologous Groups (COG), gene ontology (GO), or Kyoto Encyclopedia of Genes and Genomes (KEGG) databases. Relative expression of liver genes between diploid and triploid bighead catfish revealed more than 90% of the annotated unigenes similarly expressed, regardless of ploidy, whereas 362 upregulated and 83 downregulated with at least a twofold change in triploid relative to diploid. Quantitative real-time PCR of 15 differentially expressed growth-related genes showed consistency between the expression profiles of those genes with the results from RNA-seq analysis. Our results showed that genes in C. macrocephalus liver responded independently to triploidy with the majority showing similar expression levels between diploid and triploid (a dosage compensation phenomenon). The underlying mechanism of the varying gene expression patterns was discussed. Notably, 5 of the top 20 upregulated genes associated with stress response and thus may reflect stress caused by triploidy. The present study adds a substantial contribution to the sequence data available for C. macrocephalus and hence provides valuable resources for further studies. Furthermore, it gives information that may enhance understanding of triploid physiology.
Collapse
Affiliation(s)
- Satid Chatchaiphan
- Graduate Program in Aquaculture, The Graduate School, Kasetsart University, Chatuchak, Bangkok, 10900, Thailand
| | - Prapansak Srisapoome
- Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Chatuchak, Bangkok, 10900, Thailand
| | - Jin-Hyoung Kim
- Division of Life Sciences, Korea Polar Research Institute, Incheon, 21990, South Korea
| | - Robert H Devlin
- Fisheries and Oceans Canada, Centre for Aquaculture and Environmental Research, West Vancouver, BC, V7V1N6, Canada
| | - Uthairat Na-Nakorn
- Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Chatuchak, Bangkok, 10900, Thailand.
| |
Collapse
|
29
|
Shah NH, Wang Q, Yan Q, Karandur D, Kadlecek TA, Fallahee IR, Russ WP, Ranganathan R, Weiss A, Kuriyan J. An electrostatic selection mechanism controls sequential kinase signaling downstream of the T cell receptor. eLife 2016; 5:e20105. [PMID: 27700984 PMCID: PMC5089863 DOI: 10.7554/elife.20105] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 10/03/2016] [Indexed: 12/15/2022] Open
Abstract
The sequence of events that initiates T cell signaling is dictated by the specificities and order of activation of the tyrosine kinases that signal downstream of the T cell receptor. Using a platform that combines exhaustive point-mutagenesis of peptide substrates, bacterial surface-display, cell sorting, and deep sequencing, we have defined the specificities of the first two kinases in this pathway, Lck and ZAP-70, for the T cell receptor ζ chain and the scaffold proteins LAT and SLP-76. We find that ZAP-70 selects its substrates by utilizing an electrostatic mechanism that excludes substrates with positively-charged residues and favors LAT and SLP-76 phosphosites that are surrounded by negatively-charged residues. This mechanism prevents ZAP-70 from phosphorylating its own activation loop, thereby enforcing its strict dependence on Lck for activation. The sequence features in ZAP-70, LAT, and SLP-76 that underlie electrostatic selectivity likely contribute to the specific response of T cells to foreign antigens.
Collapse
Affiliation(s)
- Neel H Shah
- Department of Molecular and Cell Biology, University of California, Berkeley, United States
- California Institute for Quantitative Biosciences, University of California, Berkeley, United States
- Howard Hughes Medical Institute, University of California, Berkeley, United States
| | - Qi Wang
- Department of Molecular and Cell Biology, University of California, Berkeley, United States
- California Institute for Quantitative Biosciences, University of California, Berkeley, United States
- Howard Hughes Medical Institute, University of California, Berkeley, United States
| | - Qingrong Yan
- Department of Molecular and Cell Biology, University of California, Berkeley, United States
- California Institute for Quantitative Biosciences, University of California, Berkeley, United States
- Howard Hughes Medical Institute, University of California, Berkeley, United States
| | - Deepti Karandur
- Department of Molecular and Cell Biology, University of California, Berkeley, United States
- California Institute for Quantitative Biosciences, University of California, Berkeley, United States
- Howard Hughes Medical Institute, University of California, Berkeley, United States
| | - Theresa A Kadlecek
- Rosalind Russell/Ephraim P Engleman Rheumatology Research Center, Department of Medicine, University of California, San Francisco, United States
- Howard Hughes Medical Institute, University of California, San Francisco, United States
| | - Ian R Fallahee
- Department of Molecular and Cell Biology, University of California, Berkeley, United States
- California Institute for Quantitative Biosciences, University of California, Berkeley, United States
- Howard Hughes Medical Institute, University of California, Berkeley, United States
| | - William P Russ
- Green Center for Systems Biology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Rama Ranganathan
- Green Center for Systems Biology, University of Texas Southwestern Medical Center, Dallas, United States
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Arthur Weiss
- Rosalind Russell/Ephraim P Engleman Rheumatology Research Center, Department of Medicine, University of California, San Francisco, United States
- Howard Hughes Medical Institute, University of California, San Francisco, United States
| | - John Kuriyan
- Department of Molecular and Cell Biology, University of California, Berkeley, United States
- California Institute for Quantitative Biosciences, University of California, Berkeley, United States
- Howard Hughes Medical Institute, University of California, Berkeley, United States
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, United States
| |
Collapse
|
30
|
Novel Grb14-Mediated Cross Talk between Insulin and p62/Nrf2 Pathways Regulates Liver Lipogenesis and Selective Insulin Resistance. Mol Cell Biol 2016; 36:2168-81. [PMID: 27215388 DOI: 10.1128/mcb.00170-16] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/17/2016] [Indexed: 12/24/2022] Open
Abstract
A long-standing paradox in the pathophysiology of metabolic diseases is the selective insulin resistance of the liver. It is characterized by a blunted action of insulin to reduce glucose production, contributing to hyperglycemia, while de novo lipogenesis remains insulin sensitive, participating in turn to hepatic steatosis onset. The underlying molecular bases of this conundrum are not yet fully understood. Here, we established a model of selective insulin resistance in mice by silencing an inhibitor of insulin receptor catalytic activity, the growth factor receptor binding protein 14 (Grb14) in liver. Indeed, Grb14 knockdown enhanced hepatic insulin signaling but also dramatically inhibited de novo fatty acid synthesis. In the liver of obese and insulin-resistant mice, downregulation of Grb14 markedly decreased blood glucose and improved liver steatosis. Mechanistic analyses showed that upon Grb14 knockdown, the release of p62/sqstm1, a partner of Grb14, activated the transcription factor nuclear factor erythroid-2-related factor 2 (Nrf2), which in turn repressed the lipogenic nuclear liver X receptor (LXR). Our study reveals that Grb14 acts as a new signaling node that regulates lipogenesis and modulates insulin sensitivity in the liver by acting at a crossroad between the insulin receptor and the p62-Nrf2-LXR signaling pathways.
Collapse
|
31
|
Moshourab R, Palada V, Grunwald S, Grieben U, Lewin GR, Spuler S. A Molecular Signature of Myalgia in Myotonic Dystrophy 2. EBioMedicine 2016; 7:205-11. [PMID: 27322473 PMCID: PMC4909324 DOI: 10.1016/j.ebiom.2016.03.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 02/26/2016] [Accepted: 03/11/2016] [Indexed: 01/12/2023] Open
Abstract
Background Chronic muscle pain affects close to 20% of the population and is a major health burden. The underlying mechanisms of muscle pain are difficult to investigate as pain presents in patients with very diverse histories. Treatment options are therefore limited and not tailored to underlying mechanisms. To gain insight into the pathophysiology of myalgia we investigated a homogeneous group of patients suffering from myotonic dystrophy type 2 (DM2), a monogenic disorder presenting with myalgia in at least 50% of affected patients. Methods After IRB approval we performed an observational cross-sectional cohort study and recruited 42 patients with genetically confirmed DM2 plus 20 healthy age and gender matched control subjects. All participants were subjected to an extensive sensory-testing protocol. In addition, RNA sequencing was performed from 12 muscle biopsy specimens obtained from DM2 patients. Findings Clinical sensory testing as well as RNA sequencing clearly separated DM2 myalgic from non-myalgia patients and also from healthy controls. In particular pressure pain thresholds were significantly lowered for all muscles tested in myalgic DM2 patients but were not significantly different between non-myalgic patients and healthy controls. The expression of fourteen muscle expressed genes in myalgic patients was significantly up or down-regulated in myalgic compared to non-myalgic DM2 patients. Interpretation Our data support the idea that molecular changes in the muscles of DM2 patients are associated with muscle pain. Further studies should address whether muscle-specific molecular pathways play a significant role in myalgia in order to facilitate the development of mechanism-based therapeutic strategies to treat musculoskeletal pain. Funding This study was funded by the German Research Society (DFG, GK1631), KAP programme of Charité Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine. Pressure pain thresholds were reduced in myotonic dystrophy 2 (DM2) and myalgias, but not in DM2 without myalgias RNASeq from skeletal muscle differed significantly in 14 genes between DM2 with myalgias and DM2 without myalgias
Patients with a rare type of muscle dystrophy (myotonic dystrophy type 2) that is caused by a single genetic mutation usually suffer from muscle weakness and wasting but a majority also suffer from chronic muscle pain in their extremities. In our study, we found that these patients are sensitive to pressure stimuli over muscles that are not usually perceived as painful. We also identified several muscle genes that might highlight a molecular link to muscle pain. These findings call for further research to learn how and whether the genetic impairments in muscle contribute to muscle pain.
Collapse
Affiliation(s)
- Rabih Moshourab
- Molecular Physiology of Somatic Sensation, Max Delbrück Center for Molecular Medicine, Berlin, Germany; Dept. of Anesthesiology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Vinko Palada
- Muscle Research Unit, Experimental and Clinical Research Center, a joint cooperation of the Charité University Medicine Berlin and the Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Stefanie Grunwald
- Muscle Research Unit, Experimental and Clinical Research Center, a joint cooperation of the Charité University Medicine Berlin and the Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Ulrike Grieben
- Muscle Research Unit, Experimental and Clinical Research Center, a joint cooperation of the Charité University Medicine Berlin and the Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Gary R Lewin
- Molecular Physiology of Somatic Sensation, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
| | - Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center, a joint cooperation of the Charité University Medicine Berlin and the Max Delbrück Center for Molecular Medicine, Berlin, Germany.
| |
Collapse
|
32
|
Yang P, Wei J, Li W, He F, Zeng S, Zhang T, Sun Z, Cao J. High expression of growth factor receptor-bound protein 14 predicts poor prognosis for colorectal cancer patients. Biotechnol Lett 2016; 38:1043-7. [PMID: 26965150 DOI: 10.1007/s10529-016-2077-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 03/01/2016] [Indexed: 12/18/2022]
Abstract
OBJECTS To explore the roles of growth factor receptor-bound protein 14 (GRB14) in colorectal cancer (CRC) and its correlation with clinicopathological characteristics and prognosis of CRC patients. RESULTS GRB14 was localized in the cytoplasm of CRC and benign glandular epithelium cells, showing higher levels in CRC tissues compared with normal colon samples (P < 0.001). High GRB14 was associated with a high pathological grade (P = 0.045), advanced clinical stage (P = 0.018), enhanced tumor invasion (P < 0.001) and lymph node metastasis (P = 0.028). The cancer genome atlas (TCGA) mRNA sequence data showed that GRB14 was upregulated in CRC at an advanced clinical stage (P = 0.011) with enhanced tumor invasion (P < 0.001) and lymph node metastasis (P = 0.014). Kaplan-Meier survival curves revealed that CRC patients with high GRB14 levels had a shorter survival compared with those showing low GRB14 expression (P = 0.007). High GRB14 expression was an independent prognostic factor for CRC patients (HR 2.847, 95 %CI 1.058-7.659; P = 0.038). CONCLUSIONS GRB14 may be an important cancer promoter that enhances CRC progression. Upregulated GRB14 levels may predict a poor clinical outcome in CRC patients.
Collapse
Affiliation(s)
- Ping Yang
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Panfu Road No. 1, Guangzhou, 510180, China
| | - Jianchang Wei
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Panfu Road No. 1, Guangzhou, 510180, China
| | - Wanglin Li
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Panfu Road No. 1, Guangzhou, 510180, China
| | - Feng He
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Panfu Road No. 1, Guangzhou, 510180, China
| | - Shanqi Zeng
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Panfu Road No. 1, Guangzhou, 510180, China
| | - Tong Zhang
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Panfu Road No. 1, Guangzhou, 510180, China
| | - Zheng Sun
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Panfu Road No. 1, Guangzhou, 510180, China
| | - Jie Cao
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Panfu Road No. 1, Guangzhou, 510180, China.
| |
Collapse
|
33
|
Kilpeläinen TO, Carli JFM, Skowronski AA, Sun Q, Kriebel J, Feitosa MF, Hedman ÅK, Drong AW, Hayes JE, Zhao J, Pers TH, Schick U, Grarup N, Kutalik Z, Trompet S, Mangino M, Kristiansson K, Beekman M, Lyytikäinen LP, Eriksson J, Henneman P, Lahti J, Tanaka T, Luan J, Greco M FD, Pasko D, Renström F, Willems SM, Mahajan A, Rose LM, Guo X, Liu Y, Kleber ME, Pérusse L, Gaunt T, Ahluwalia TS, Ju Sung Y, Ramos YF, Amin N, Amuzu A, Barroso I, Bellis C, Blangero J, Buckley BM, Böhringer S, I Chen YD, de Craen AJN, Crosslin DR, Dale CE, Dastani Z, Day FR, Deelen J, Delgado GE, Demirkan A, Finucane FM, Ford I, Garcia ME, Gieger C, Gustafsson S, Hallmans G, Hankinson SE, Havulinna AS, Herder C, Hernandez D, Hicks AA, Hunter DJ, Illig T, Ingelsson E, Ioan-Facsinay A, Jansson JO, Jenny NS, Jørgensen ME, Jørgensen T, Karlsson M, Koenig W, Kraft P, Kwekkeboom J, Laatikainen T, Ladwig KH, LeDuc CA, Lowe G, Lu Y, Marques-Vidal P, Meisinger C, Menni C, Morris AP, Myers RH, Männistö S, Nalls MA, Paternoster L, Peters A, Pradhan AD, Rankinen T, Rasmussen-Torvik LJ, Rathmann W, Rice TK, Brent Richards J, Ridker PM, Sattar N, Savage DB, et alKilpeläinen TO, Carli JFM, Skowronski AA, Sun Q, Kriebel J, Feitosa MF, Hedman ÅK, Drong AW, Hayes JE, Zhao J, Pers TH, Schick U, Grarup N, Kutalik Z, Trompet S, Mangino M, Kristiansson K, Beekman M, Lyytikäinen LP, Eriksson J, Henneman P, Lahti J, Tanaka T, Luan J, Greco M FD, Pasko D, Renström F, Willems SM, Mahajan A, Rose LM, Guo X, Liu Y, Kleber ME, Pérusse L, Gaunt T, Ahluwalia TS, Ju Sung Y, Ramos YF, Amin N, Amuzu A, Barroso I, Bellis C, Blangero J, Buckley BM, Böhringer S, I Chen YD, de Craen AJN, Crosslin DR, Dale CE, Dastani Z, Day FR, Deelen J, Delgado GE, Demirkan A, Finucane FM, Ford I, Garcia ME, Gieger C, Gustafsson S, Hallmans G, Hankinson SE, Havulinna AS, Herder C, Hernandez D, Hicks AA, Hunter DJ, Illig T, Ingelsson E, Ioan-Facsinay A, Jansson JO, Jenny NS, Jørgensen ME, Jørgensen T, Karlsson M, Koenig W, Kraft P, Kwekkeboom J, Laatikainen T, Ladwig KH, LeDuc CA, Lowe G, Lu Y, Marques-Vidal P, Meisinger C, Menni C, Morris AP, Myers RH, Männistö S, Nalls MA, Paternoster L, Peters A, Pradhan AD, Rankinen T, Rasmussen-Torvik LJ, Rathmann W, Rice TK, Brent Richards J, Ridker PM, Sattar N, Savage DB, Söderberg S, Timpson NJ, Vandenput L, van Heemst D, Uh HW, Vohl MC, Walker M, Wichmann HE, Widén E, Wood AR, Yao J, Zeller T, Zhang Y, Meulenbelt I, Kloppenburg M, Astrup A, Sørensen TIA, Sarzynski MA, Rao DC, Jousilahti P, Vartiainen E, Hofman A, Rivadeneira F, Uitterlinden AG, Kajantie E, Osmond C, Palotie A, Eriksson JG, Heliövaara M, Knekt PB, Koskinen S, Jula A, Perola M, Huupponen RK, Viikari JS, Kähönen M, Lehtimäki T, Raitakari OT, Mellström D, Lorentzon M, Casas JP, Bandinelli S, März W, Isaacs A, van Dijk KW, van Duijn CM, Harris TB, Bouchard C, Allison MA, Chasman DI, Ohlsson C, Lind L, Scott RA, Langenberg C, Wareham NJ, Ferrucci L, Frayling TM, Pramstaller PP, Borecki IB, Waterworth DM, Bergmann S, Waeber G, Vollenweider P, Vestergaard H, Hansen T, Pedersen O, Hu FB, Eline Slagboom P, Grallert H, Spector TD, Jukema J, Klein RJ, Schadt EE, Franks PW, Lindgren CM, Leibel RL, Loos RJF. Genome-wide meta-analysis uncovers novel loci influencing circulating leptin levels. Nat Commun 2016; 7:10494. [PMID: 26833098 PMCID: PMC4740377 DOI: 10.1038/ncomms10494] [Show More Authors] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 12/16/2015] [Indexed: 01/20/2023] Open
Abstract
Leptin is an adipocyte-secreted hormone, the circulating levels of which correlate closely with overall adiposity. Although rare mutations in the leptin (LEP) gene are well known to cause leptin deficiency and severe obesity, no common loci regulating circulating leptin levels have been uncovered. Therefore, we performed a genome-wide association study (GWAS) of circulating leptin levels from 32,161 individuals and followed up loci reaching P<10(-6) in 19,979 additional individuals. We identify five loci robustly associated (P<5 × 10(-8)) with leptin levels in/near LEP, SLC32A1, GCKR, CCNL1 and FTO. Although the association of the FTO obesity locus with leptin levels is abolished by adjustment for BMI, associations of the four other loci are independent of adiposity. The GCKR locus was found associated with multiple metabolic traits in previous GWAS and the CCNL1 locus with birth weight. Knockdown experiments in mouse adipose tissue explants show convincing evidence for adipogenin, a regulator of adipocyte differentiation, as the novel causal gene in the SLC32A1 locus influencing leptin levels. Our findings provide novel insights into the regulation of leptin production by adipose tissue and open new avenues for examining the influence of variation in leptin levels on adiposity and metabolic health.
Collapse
Affiliation(s)
- Tuomas O. Kilpeläinen
- The Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, Universitetsparken 1, DIKU
Building, Copenhagen
2100, Denmark
- MRC Epidemiology Unit, Institute of Metabolic Science,
University of Cambridge, Cambridge
CB2 0QQ, UK
- Genetics of Obesity and Related Metabolic Traits Program,
Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine
at Mount Sinai, New York, New York
10029, USA
| | - Jayne F. Martin Carli
- Department of Biochemistry and Molecular Biophysics, Columbia
University, New York, New York
10032, USA
| | - Alicja A. Skowronski
- Institute of Human Nutrition, Columbia University,
New York, New York
10032, USA
| | - Qi Sun
- Channing Division of Network Medicine, Department of Medicine,
Brigham and Women's Hospital and Harvard Medical School,
Boston, Massachussetts
02115, USA
- Department of Nutrition, Harvard T.H. Chan School of Public
Health, Boston, Massachussetts
02115, USA
| | - Jennifer Kriebel
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum
München - German Research Center for Environmental Health,
Neuherberg
85764, Germany
- Institute of Epidemiology II, Helmholtz Zentrum
München-German Research Center for Environmental Health,
Neuherberg
85764, Germany
- German Center for Diabetes Research (DZD),
München-Neuherberg
85764, Germany
| | - Mary F Feitosa
- Department of Genetics, Washington University School of
Medicine, St. Louis, Missouri
63110, USA
| | - Åsa K. Hedman
- Science for Life Laboratory, Uppsala University,
Uppsala
750 85, Sweden
- Department of Medical Sciences, Molecular Epidemiology, Uppsala
University, Uppsala
751 85, Sweden
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
| | - Alexander W. Drong
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
| | - James E. Hayes
- Cell and Developmental Biology Graduate Program, Weill Cornell
Graduate School of Medical Sciences, Cornell University, New
York, New York
10021, USA
- Icahn Institute for Genomics and Multiscale Biology, Icahn
School of Medicine at Mount Sinai, New York, New York
10029, USA
| | - Jinghua Zhao
- MRC Epidemiology Unit, Institute of Metabolic Science,
University of Cambridge, Cambridge
CB2 0QQ, UK
| | - Tune H. Pers
- The Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, Universitetsparken 1, DIKU
Building, Copenhagen
2100, Denmark
- Divisions of Endocrinology and Genetics and Center for Basic
and Translational Obesity Research, Boston Children's Hospital,
Boston, Massachussetts
02115, USA
- Broad Institute of the Massachusetts Institute of Technology
and Harvard University, Cambridge, Massachusetts
2142, USA
- Department of Genetics, Harvard Medical School,
Boston, Massachusetts
02115, USA
- Department of Epidemiology Research, Statens Serum
Institut, Copenhagen
2300, Denmark
| | - Ursula Schick
- Genetics of Obesity and Related Metabolic Traits Program,
Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine
at Mount Sinai, New York, New York
10029, USA
| | - Niels Grarup
- The Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, Universitetsparken 1, DIKU
Building, Copenhagen
2100, Denmark
| | - Zoltán Kutalik
- Institute of Social and Preventive Medicine, Lausanne
University Hospital, Lausanne
1010, Switzerland
- Swiss Institute of Bioinformatics, Lausanne
1015, Switzerland
| | - Stella Trompet
- Department of Cardiology, Leiden University Medical
Center, Leiden
2333, The Netherlands
- Department of Gerontology and Geriatrics, Leiden University
Medical Center, Leiden
2333, The Netherlands
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology,
King's College London, London
SE1 7EH, UK
- National Institute for Health Research Biomedical Research
Centre at Guy's and St. Thomas' Foundation Trust,
London
SE1 9RT, UK
| | - Kati Kristiansson
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
- Institute for Molecular Medicine Finland, University of
Helsinki, Helsinki
FI-00290, Finland
| | - Marian Beekman
- Department of Molecular Epidemiology, Leiden University Medical
Center, Leiden
2300 RC, The Netherlands
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories,
Tampere
FI-33101, Finland
- Department of Clinical Chemistry, University of Tampere School
of Medicine, Tampere
FI-33014, Finland
| | - Joel Eriksson
- Centre for Bone and Arthritis Research, Department of Internal
Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy,
University of Gothenburg, Gothenburg
413 45, Sweden
| | - Peter Henneman
- Department of Human Genetics, Leiden University Medical
Center, Leiden
2333, The Netherlands
- Department of Clinical Genetics, Amsterdam Medical
Center, Amsterdam
1081 HV, The Netherlands
| | - Jari Lahti
- Institute of Behavioural Sciences, University of
Helsinki, Helsinki
FI-00014, Finland
- Folkhälsan Research Center, Helsinki
FI-00290, Finland
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on
Aging, Baltimore, Maryland
21225, USA
| | - Jian'an Luan
- MRC Epidemiology Unit, Institute of Metabolic Science,
University of Cambridge, Cambridge
CB2 0QQ, UK
| | - Fabiola Del Greco M
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC)
- Affiliated Institute of the University of Lübeck,
Bolzano
39100, Italy
| | - Dorota Pasko
- Genetics of Complex Traits, University of Exeter Medical
School, University of Exeter, Exeter
EX2 5DW, UK
| | - Frida Renström
- Department of Clinical Sciences, Genetic and Molecular
Epidemiology Unit, Lund University, Malmö
20502, Sweden
- Department of Biobank Research, Umeå
University, Umeå
90187, Sweden
| | - Sara M. Willems
- Department of Epidemiology, Erasmus MC,
Rotterdam
3015 GE, The Netherlands
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
| | - Lynda M. Rose
- Division of Preventive Medicine, Brigham and Women's
Hospital, Boston, Massachussetts
02215, USA
| | - Xiuqing Guo
- Department of Pediatrics, LABioMed at Harbor-UCLA Medical
Center, Institute for Translational Genomics and Population Sciences,
Torrance, California
90502, USA
| | - Yongmei Liu
- Center for Human Genetics, Division of Public Health Sciences,
Wake Forest School of Medicine, Winston-Salem, North
Carolina
27157, USA
| | - Marcus E. Kleber
- Medical Faculty Mannheim, Vth Department of Medicine,
Heidelberg University, Mannheim
68167, Germany
| | - Louis Pérusse
- Department of Kinesiology, Laval University, Quebec
City, Quebec, Canada
G1V 0A6
- Institute of Nutrition and Functional Foods, Quebec
City, Quebec, Canada
G1V 0A6
| | - Tom Gaunt
- MRC Integrative Epidemiology Unit and School of Social and
Community Medicine, University of Bristol, Bristol
BS82BN, UK
| | - Tarunveer S. Ahluwalia
- The Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, Universitetsparken 1, DIKU
Building, Copenhagen
2100, Denmark
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood,
Herlev and Gentofte Hospital, University of Copenhagen, Ledreborg
Allé, Copenhagen
DK-2820, Denmark
- Steno Diabetes Center, Gentofte
DK-2820, Denmark
| | - Yun Ju Sung
- Division of Biostatistics, Washington University School of
Medicine, St. Louis, Missouri
63108, USA
- Department of Psychiatry, Washington University School of
Medicine, St. Louis, Missouri
63110, USA
| | - Yolande F. Ramos
- Department of Molecular Epidemiology, Leiden University Medical
Center, Leiden
2300 RC, The Netherlands
| | - Najaf Amin
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus
MC, Rotterdam
3015 GE, The Netherlands
| | - Antoinette Amuzu
- Faculty of Epidemiology and Population Health, London School of
Hygiene and Tropical Medicine, London
WC1E 7HT, UK
| | - Inês Barroso
- Wellcome Trust Sanger Institute, Hinxton
CB10 1SA, UK
- NIHR Cambridge Biomedical Research Centre, Institute of
Metabolic Science, Addenbrooke's Hospital, Cambridge
CB2 0QQ, UK
- The University of Cambridge Metabolic Research Laboratories,
Wellcome Trust-MRC Institute of Metabolic Science, Cambridge
CB2 0QQ, UK
| | - Claire Bellis
- Human Genetics, Genome Institute of Singapore, Agency for
Science, Technology and Research of Singapore, Singapore
138672, Singapore
- Genomics Research Centre, Institute of Health and Biomedical
Innovation, Queensland University of Technology, Brisbane,
Queensland
4001, Australia
- Texas Biomedical Research Institute, San
Antonio, Texas
78245, USA
| | - John Blangero
- Texas Biomedical Research Institute, San
Antonio, Texas
78245, USA
| | - Brendan M. Buckley
- Department of Pharmacology and Therapeutics, University College
Cork, Cork
T12 YT57, Ireland
| | - Stefan Böhringer
- Department of Molecular Epidemiology, Leiden University Medical
Center, Leiden
2300 RC, The Netherlands
| | - Yii-Der I Chen
- Department of Pediatrics, LABioMed at Harbor-UCLA Medical
Center, Institute for Translational Genomics and Population Sciences,
Torrance, California
90502, USA
| | - Anton J. N. de Craen
- Department of Gerontology and Geriatrics, Leiden University
Medical Center, Leiden
2333, The Netherlands
| | - David R. Crosslin
- Division of Medical Genetics, Department of Medicine,
University of Washington, Seattle, Washington
98195, USA
- Department of Genome Sciences, University of Washington,
Seattle, Washington
98195, USA
| | - Caroline E. Dale
- Faculty of Epidemiology and Population Health, London School of
Hygiene and Tropical Medicine, London
WC1E 7HT, UK
| | - Zari Dastani
- Department of Human Genetics, McGill University,
Montreal, Quebec, Canada
H3A 0G4
| | - Felix R. Day
- MRC Epidemiology Unit, Institute of Metabolic Science,
University of Cambridge, Cambridge
CB2 0QQ, UK
| | - Joris Deelen
- Department of Molecular Epidemiology, Leiden University Medical
Center, Leiden
2300 RC, The Netherlands
| | - Graciela E. Delgado
- Medical Faculty Mannheim, Vth Department of Medicine,
Heidelberg University, Mannheim
68167, Germany
| | - Ayse Demirkan
- Department of Human Genetics, Leiden University Medical
Center, Leiden
2333, The Netherlands
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus
MC, Rotterdam
3015 GE, The Netherlands
| | - Francis M. Finucane
- MRC Epidemiology Unit, Institute of Metabolic Science,
University of Cambridge, Cambridge
CB2 0QQ, UK
| | - Ian Ford
- Robertson Center for Biostatistics, University of
Glasgow, Glasgow
G12 8QQ, UK
| | - Melissa E. Garcia
- National Heart, Lung, and Blood Institute, NIH,
Bethesda, Maryland
2089, USA
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum
München - German Research Center for Environmental Health,
Neuherberg
85764, Germany
- Institute of Epidemiology II, Helmholtz Zentrum
München-German Research Center for Environmental Health,
Neuherberg
85764, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum
München, German Research Center for Environmental Health,
Neuherberg
85764, Germany
| | - Stefan Gustafsson
- Science for Life Laboratory, Uppsala University,
Uppsala
750 85, Sweden
- Department of Medical Sciences, Molecular Epidemiology, Uppsala
University, Uppsala
751 85, Sweden
| | - Göran Hallmans
- Department of Biobank Research, Umeå
University, Umeå
90187, Sweden
| | - Susan E. Hankinson
- Channing Division of Network Medicine, Department of Medicine,
Brigham and Women's Hospital and Harvard Medical School,
Boston, Massachussetts
02115, USA
- Department of Biostatistics and Epidemiology, School of Public
Health and Health Sciences, University of Massachusetts,
Amherst, Massachusetts
01003, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public
Health, Boston, Massachusetts
02115, USA
| | - Aki S Havulinna
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
| | - Christian Herder
- German Center for Diabetes Research (DZD),
München-Neuherberg
85764, Germany
- Institute for Clinical Diabetology, German Diabetes Center,
Leibniz Center for Diabetes Research at Heinrich Heine University
Düsseldorf, Düsseldorf
40225, Germany
| | - Dena Hernandez
- Laboratory of Neurogenetics, National Institute on Aging,
Bethesda, Maryland
20892, USA
| | - Andrew A. Hicks
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC)
- Affiliated Institute of the University of Lübeck,
Bolzano
39100, Italy
| | - David J. Hunter
- Department of Nutrition and Epidemiology, Harvard T.H. Chan
School of Public Health, Boston, Massachusetts
02115, USA
| | - Thomas Illig
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum
München - German Research Center for Environmental Health,
Neuherberg
85764, Germany
- Hannover Unified Biobank, Hannover Medical School,
Hannover
30625, Germany
- Institute for Human Genetics, Hannover Medical School,
Hannover
30625, Germany
| | - Erik Ingelsson
- Science for Life Laboratory, Uppsala University,
Uppsala
750 85, Sweden
- Department of Medical Sciences, Molecular Epidemiology, Uppsala
University, Uppsala
751 85, Sweden
- Division of Cardiovascular Medicine, Department of Medicine,
Stanford University School of Medicine, Stanford,
California
94305, USA
| | - Andreea Ioan-Facsinay
- Department of Rheumatology, Leiden University Medical
Center, Leiden
2333, The Netherlands
| | - John-Olov Jansson
- Department of Physiology, Institute of Neuroscience and
Physiology, Sahlgrenska Academy, University of Gothenburg,
Gothenburg
41345, Sweden
| | - Nancy S. Jenny
- Laboratory for Clinical Biochemistry Research, Department of
Pathology and Laboratory Medicine, University of Vermont College of
Medicine, Colchester, Vermont
05405, USA
| | | | - Torben Jørgensen
- Research Centre for Prevention and Health, Glostrup University
Hospital, Glostrup
2600, Denmark
- Faculty of Medicine, University of Aalborg,
Aalborg
9100, Denmark
- Faculty of Health and Medical Sciences, University of
Copenhagen, Copenhagen
2200, Denmark
| | - Magnus Karlsson
- Clinical and Molecular Osteoporosis Research Unit, Department
of Clinical Sciences and Orthopaedic Surgery, Lund University, Skåne
University Hospital, Malmö
21428, Sweden
| | - Wolfgang Koenig
- Department of Internal Medicine II - Cardiology, University of
Ulm, Ulm
89081, Germany
- Deutsches Herzzentrum München, Technische
Universität München, Munich
80636, Germany
- DZHK (German Centre for Cardiovascular Research), partner site
Munich Heart Alliance, Munich
80539, Germany
| | - Peter Kraft
- Department of Epidemiology and Biostatistics, Harvard T.H. Chan
School of Public Health, Boston, Massachussetts
02115, USA
| | - Joanneke Kwekkeboom
- Department of Rheumatology, Leiden University Medical
Center, Leiden
2333, The Netherlands
| | - Tiina Laatikainen
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
- Institute of Public Health and Clinical Nutrition, University
of Eastern Finland, Kuopio
FI-70211, Finland
- Hospital District of North Karelia, Joensuu
FI-80210, Finland
| | - Karl-Heinz Ladwig
- Institute of Epidemiology II, Helmholtz Zentrum
München-German Research Center for Environmental Health,
Neuherberg
85764, Germany
- Department of Psychosomatic Medicine and Psychotherapy,
Klinikum Rechts der Isar, Technische Universität
München, Munich
81675, Germany
| | - Charles A. LeDuc
- Division of Molecular Genetics, Department of Pediatrics,
Columbia University, New York, New York
10029, USA
| | - Gordon Lowe
- Institute of Cardiovascular and Medical Sciences, University of
Glasgow, Glasgow
G12 8QQ, UK
| | - Yingchang Lu
- Genetics of Obesity and Related Metabolic Traits Program,
Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine
at Mount Sinai, New York, New York
10029, USA
| | - Pedro Marques-Vidal
- Department of Internal Medicine, Lausanne University
Hospital, Lausanne
1011, Switzerland
| | - Christa Meisinger
- Institute of Epidemiology II, Helmholtz Zentrum
München-German Research Center for Environmental Health,
Neuherberg
85764, Germany
- German Center for Diabetes Research (DZD),
München-Neuherberg
85764, Germany
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology,
King's College London, London
SE1 7EH, UK
| | - Andrew P. Morris
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
- Department of Biostatistics, University of Liverpool,
Liverpool
L69 3GA, UK
| | - Richard H. Myers
- Department of Neurology, Boston University School of
Medicine, Boston, Massachussetts
02118, USA
| | - Satu Männistö
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
| | - Mike A. Nalls
- Laboratory of Neurogenetics, National Institute on Aging,
Bethesda, Maryland
20892, USA
| | - Lavinia Paternoster
- MRC Integrative Epidemiology Unit and School of Social and
Community Medicine, University of Bristol, Bristol
BS82BN, UK
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum
München-German Research Center for Environmental Health,
Neuherberg
85764, Germany
- German Center for Diabetes Research (DZD),
München-Neuherberg
85764, Germany
- DZHK (German Centre for Cardiovascular Research), partner site
Munich Heart Alliance, Munich
80539, Germany
| | - Aruna D. Pradhan
- Division of Preventive Medicine, Brigham and Women's
Hospital, Boston, Massachussetts
02215, USA
- Harvard Medical School, Boston,
Massachussetts
02115, USA
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research
Center, Baton Rouge, Los Angeles
70808, USA
| | | | - Wolfgang Rathmann
- Institute for Biometrics and Epidemiology, German Diabetes
Center, Leibniz Center for Diabetes Research at Heinrich Heine University
Düsseldorf, Düsseldorf
40225, Germany
| | - Treva K. Rice
- Division of Biostatistics, Washington University School of
Medicine, St. Louis, Missouri
63108, USA
- Department of Psychiatry, Washington University School of
Medicine, St. Louis, Missouri
63110, USA
| | - J Brent Richards
- Department of Twin Research and Genetic Epidemiology,
King's College London, London
SE1 7EH, UK
- Department of Medicine, Human Genetics and Epidemiology,
McGill University, Montreal, Quebec, Canada
H3A 0G4
| | - Paul M. Ridker
- Division of Preventive Medicine, Brigham and Women's
Hospital, Boston, Massachussetts
02215, USA
- Harvard Medical School, Boston,
Massachussetts
02115, USA
| | - Naveed Sattar
- Faculty of Medicine, BHF Glasgow Cardiovascular Research
Centre, Glasgow
G12 8QQ, UK
| | - David B. Savage
- The University of Cambridge Metabolic Research Laboratories,
Wellcome Trust-MRC Institute of Metabolic Science, Cambridge
CB2 0QQ, UK
| | - Stefan Söderberg
- Department of Public Health and Clinical Medicine, Cardiology
and Heart Centre, Umeå University, Umeå
90187, Sweden
| | - Nicholas J. Timpson
- MRC Integrative Epidemiology Unit and School of Social and
Community Medicine, University of Bristol, Bristol
BS82BN, UK
| | - Liesbeth Vandenput
- Centre for Bone and Arthritis Research, Department of Internal
Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy,
University of Gothenburg, Gothenburg
413 45, Sweden
| | - Diana van Heemst
- Department of Gerontology and Geriatrics, Leiden University
Medical Center, Leiden
2333, The Netherlands
| | - Hae-Won Uh
- Department of Molecular Epidemiology, Leiden University Medical
Center, Leiden
2300 RC, The Netherlands
| | - Marie-Claude Vohl
- Institute of Nutrition and Functional Foods, Quebec
City, Quebec, Canada
G1V 0A6
- School of Nutrition, Laval University, Quebec
City, Quebec, Canada
G1V 0A6
| | - Mark Walker
- Institute of Cellular Medicine, Newcastle University,
Newcastle upon Tyne
NE1 7RU, UK
| | - Heinz-Erich Wichmann
- Institute of Medical Informatics, Biometry and Epidemiology,
Ludwig-Maximilians-Universität and Klinikum Grosshadern,
Munich
80336, Germany
- Institute of Epidemiology I, Helmholtz Zentrum
München-German Research Center for Environmental Health,
Neuherberg
85764, Germany
- Institute of Medical Statistics and Epidemiology, Technical
University Munich, Munich
81675, Germany
| | - Elisabeth Widén
- Institute for Molecular Medicine Finland, University of
Helsinki, Helsinki
FI-00290, Finland
| | - Andrew R. Wood
- Genetics of Complex Traits, University of Exeter Medical
School, University of Exeter, Exeter
EX2 5DW, UK
| | - Jie Yao
- Department of Pediatrics, LABioMed at Harbor-UCLA Medical
Center, Institute for Translational Genomics and Population Sciences,
Torrance, California
90502, USA
| | - Tanja Zeller
- German Center for Cardiovascular Research (DZHK e.V.), partner
site Hamburg/Kiel/Lübeck, Hamburg
20246, Germany
- Clinic for General and Interventional Cardiology, University
Heart Center Hamburg, Hamburg
20246, Germany
| | - Yiying Zhang
- Division of Molecular Genetics, Department of Pediatrics,
Columbia University, New York, New York
10029, USA
| | - Ingrid Meulenbelt
- Department of Molecular Epidemiology, Leiden University Medical
Center, Leiden
2300 RC, The Netherlands
| | - Margreet Kloppenburg
- Department of Rheumatology, Leiden University Medical
Center, Leiden
2333, The Netherlands
- Department of Clinical Epidemiology, Leiden University Medical
Center, Leiden
2333, The Netherlands
| | - Arne Astrup
- Faculty of Science, Department of Nutrition, Exercise, and
Sports, University of Copenhagen, Copenhagen 1165, Denmark
| | - Thorkild I. A. Sørensen
- The Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, Universitetsparken 1, DIKU
Building, Copenhagen
2100, Denmark
- MRC Integrative Epidemiology Unit and School of Social and
Community Medicine, University of Bristol, Bristol
BS82BN, UK
- Institute of Preventive Medicine, Bispebjerg and Frederiksberg
Hospitals, The Capital Region, Copenhagen
2000, Denmark
| | - Mark A. Sarzynski
- Human Genomics Laboratory, Pennington Biomedical Research
Center, Baton Rouge, Los Angeles
70808, USA
| | - D. C. Rao
- Department of Genetics, Washington University School of
Medicine, St. Louis, Missouri
63110, USA
- Division of Biostatistics, Washington University School of
Medicine, St. Louis, Missouri
63108, USA
- Department of Psychiatry, Washington University School of
Medicine, St. Louis, Missouri
63110, USA
| | - Pekka Jousilahti
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
| | - Erkki Vartiainen
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
| | - Albert Hofman
- Department of Epidemiology, Erasmus MC,
Rotterdam
3015 GE, The Netherlands
| | - Fernando Rivadeneira
- Department of Epidemiology, Erasmus MC,
Rotterdam
3015 GE, The Netherlands
- Department of Internal Medicine, Erasmus MC,
Rotterdam
3015 GE, The Netherlands
| | - André G. Uitterlinden
- Department of Epidemiology, Erasmus MC,
Rotterdam
3015 GE, The Netherlands
- Department of Internal Medicine, Erasmus MC,
Rotterdam
3015 GE, The Netherlands
| | - Eero Kajantie
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
- Children's Hospital, Helsinki University Central
Hospital and University of Helsinki, Helsinki
FI-00014, Finland
- Department of Obstetrics and Gynaecology, MRC Oulu, Oulu
University Central Hospital and University of Oulu, Oulu
90220, Finland
| | - Clive Osmond
- MRC Lifecourse Epidemiology Unit, University of Southampton,
Southampton General Hospital, Southampton
SO16 6YD, UK
| | - Aarno Palotie
- Institute for Molecular Medicine Finland, University of
Helsinki, Helsinki
FI-00290, Finland
- Wellcome Trust Sanger Institute, Hinxton
CB10 1SA, UK
- Center for Human Genetic Research, Psychiatric and
Neurodevelopmental Genetics Unit, Massachusetts General Hospital,
Boston, Massachusetts
02114, USA
| | - Johan G. Eriksson
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
- Folkhälsan Research Center, Helsinki
FI-00290, Finland
- Department of General Practice and Primary Health Care,
University of Helsinki, Helsinki
FI-00014, Finland
| | - Markku Heliövaara
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
| | - Paul B. Knekt
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
| | - Seppo Koskinen
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
| | - Antti Jula
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
| | - Markus Perola
- Department of Health, National Institute for Health and
Welfare, Helsinki
FI-00271, Finland
- Institute for Molecular Medicine Finland, University of
Helsinki, Helsinki
FI-00290, Finland
- University of Tartu, Estonian Genome Centre,
Tartu
51010, Estonia
| | - Risto K. Huupponen
- Department of Pharmacology, Drug Development and Therapeutics,
University of Turku, Turku
FI-20520, Finland
- Unit of Clinical Pharmacology, Turku University
Hospital, Turku
FI-20520, Finland
| | - Jorma S. Viikari
- Division of Medicine, Turku University Hospital,
Turku
FI-20520, Finland
- Department of Medicine, University of Turku,
Turku
FI-20520, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University
Hospital, Tampere
FI-33521, Finland
- Department of Clinical Physiology, University of Tampere
School of Medicine, Tampere
FI-33014, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories,
Tampere
FI-33101, Finland
- Department of Clinical Chemistry, University of Tampere School
of Medicine, Tampere
FI-33014, Finland
| | - Olli T. Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku
University Hospital, Turku
FI-2051, Finland
- Research Centre of Applied and Preventive Cardiovascular
Medicine, University of Turku, Turku
FI-20520, Finland
| | - Dan Mellström
- Centre for Bone and Arthritis Research, Department of Internal
Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy,
University of Gothenburg, Gothenburg
413 45, Sweden
| | - Mattias Lorentzon
- Centre for Bone and Arthritis Research, Department of Internal
Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy,
University of Gothenburg, Gothenburg
413 45, Sweden
| | - Juan P. Casas
- Farr Institute of Health Informatics, University College
London, London
NW1 2DA, UK
| | | | - Winfried März
- Medical Faculty Mannheim, Vth Department of Medicine,
Heidelberg University, Mannheim
68167, Germany
- Synlab Academy, Synlab Services LLC, Mannheim
68161, Germany
- Clinical Institute of Medical and Chemical Laboratory
Diagnostics, Medical University of Graz, Graz
8010, Austria
| | - Aaron Isaacs
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus
MC, Rotterdam
3015 GE, The Netherlands
| | - Ko W. van Dijk
- Department of Human Genetics, Leiden University Medical
Center, Leiden
2333, The Netherlands
| | - Cornelia M. van Duijn
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus
MC, Rotterdam
3015 GE, The Netherlands
- Center of Medical Systems Biology, Leiden
2300 RC, The Netherlands
| | - Tamara B. Harris
- Laboratory of Epidemiology and Population Science, National
Institute on Aging, Bethesda, Maryland
20892, USA
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research
Center, Baton Rouge, Los Angeles
70808, USA
| | - Matthew A. Allison
- Family and Preventive Medicine, University of
California–San Diego, La Jolla, California
92161, USA
| | - Daniel I. Chasman
- Division of Preventive Medicine, Brigham and Women's
Hospital, Boston, Massachussetts
02215, USA
- Harvard Medical School, Boston,
Massachussetts
02115, USA
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Department of Internal
Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy,
University of Gothenburg, Gothenburg
413 45, Sweden
| | - Lars Lind
- Department of Medical Sciences, Cardiovascular Epidemiology,
Uppsala University, Uppsala
75185, Sweden
| | - Robert A. Scott
- MRC Epidemiology Unit, Institute of Metabolic Science,
University of Cambridge, Cambridge
CB2 0QQ, UK
| | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science,
University of Cambridge, Cambridge
CB2 0QQ, UK
| | - Nicholas J. Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science,
University of Cambridge, Cambridge
CB2 0QQ, UK
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on
Aging, Baltimore, Maryland
21225, USA
| | - Timothy M. Frayling
- Genetics of Complex Traits, University of Exeter Medical
School, University of Exeter, Exeter
EX2 5DW, UK
| | - Peter P. Pramstaller
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC)
- Affiliated Institute of the University of Lübeck,
Bolzano
39100, Italy
- Department of Neurology, General Central Hospital,
Bolzano
39100, Italy
- Department of Neurology, University of Lübeck,
Lübeck
23562, Germany
| | - Ingrid B. Borecki
- Department of Genetics, Washington University School of
Medicine, St. Louis, Missouri
63110, USA
| | | | - Sven Bergmann
- Swiss Institute of Bioinformatics, Lausanne
1015, Switzerland
- Department of Medical Genetics, University of Lausanne,
Lausanne
1015, Switzerland
| | - Gérard Waeber
- Department of Internal Medicine, Lausanne University
Hospital, Lausanne
1011, Switzerland
| | - Peter Vollenweider
- Department of Internal Medicine, Lausanne University
Hospital, Lausanne
1011, Switzerland
| | - Henrik Vestergaard
- The Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, Universitetsparken 1, DIKU
Building, Copenhagen
2100, Denmark
- Steno Diabetes Center, Gentofte
DK-2820, Denmark
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, Universitetsparken 1, DIKU
Building, Copenhagen
2100, Denmark
- Faculty of Health Sciences, University of Southern
Denmark, Odense
5230, Denmark
| | - Oluf Pedersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, Universitetsparken 1, DIKU
Building, Copenhagen
2100, Denmark
| | - Frank B. Hu
- Department of Nutrition and Epidemiology, Harvard T.H. Chan
School of Public Health, Boston, Massachusetts
02115, USA
| | - P Eline Slagboom
- Department of Molecular Epidemiology, Leiden University Medical
Center, Leiden
2300 RC, The Netherlands
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum
München - German Research Center for Environmental Health,
Neuherberg
85764, Germany
- Institute of Epidemiology II, Helmholtz Zentrum
München-German Research Center for Environmental Health,
Neuherberg
85764, Germany
- German Center for Diabetes Research (DZD),
München-Neuherberg
85764, Germany
| | - Tim D. Spector
- Department of Twin Research and Genetic Epidemiology,
King's College London, London
SE1 7EH, UK
| | - J.W. Jukema
- Department of Cardiology, Leiden University Medical
Center, Leiden
2333, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands,
Utrecht
3511 EP, The Netherlands
- Durrer Center for Cardiogenetic Research,
Amsterdam
1105 AZ, The Netherlands
| | - Robert J. Klein
- Icahn Institute for Genomics and Multiscale Biology, Icahn
School of Medicine at Mount Sinai, New York, New York
10029, USA
| | - Erik E Schadt
- Icahn Institute for Genomics and Multiscale Biology, Icahn
School of Medicine at Mount Sinai, New York, New York
10029, USA
| | - Paul W. Franks
- Department of Nutrition, Harvard T.H. Chan School of Public
Health, Boston, Massachussetts
02115, USA
- Department of Clinical Sciences, Genetic and Molecular
Epidemiology Unit, Lund University, Malmö
20502, Sweden
- Department of Public Health and Clinical Medicine,
Umeå University, Umeå
90187, Sweden
| | - Cecilia M. Lindgren
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
- Program in Medical and Population Genetics, Broad
Institute, Cambridge, Massachussetts
02142, USA
- The Big Data Institute, University of Oxford,
Oxford
OX1 2JD, UK
| | - Rudolph L. Leibel
- Division of Molecular Genetics, Department of Pediatrics,
Columbia University, New York, New York
10029, USA
| | - Ruth J. F. Loos
- MRC Epidemiology Unit, Institute of Metabolic Science,
University of Cambridge, Cambridge
CB2 0QQ, UK
- Genetics of Obesity and Related Metabolic Traits Program,
Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine
at Mount Sinai, New York, New York
10029, USA
- The Mindich Child Health and Development Institute, Icahn
School of Medicine at Mount Sinai, New York, New York
10029, USA
| |
Collapse
|
34
|
Lu S, Jang H, Muratcioglu S, Gursoy A, Keskin O, Nussinov R, Zhang J. Ras Conformational Ensembles, Allostery, and Signaling. Chem Rev 2016; 116:6607-65. [PMID: 26815308 DOI: 10.1021/acs.chemrev.5b00542] [Citation(s) in RCA: 283] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Ras proteins are classical members of small GTPases that function as molecular switches by alternating between inactive GDP-bound and active GTP-bound states. Ras activation is regulated by guanine nucleotide exchange factors that catalyze the exchange of GDP by GTP, and inactivation is terminated by GTPase-activating proteins that accelerate the intrinsic GTP hydrolysis rate by orders of magnitude. In this review, we focus on data that have accumulated over the past few years pertaining to the conformational ensembles and the allosteric regulation of Ras proteins and their interpretation from our conformational landscape standpoint. The Ras ensemble embodies all states, including the ligand-bound conformations, the activated (or inactivated) allosteric modulated states, post-translationally modified states, mutational states, transition states, and nonfunctional states serving as a reservoir for emerging functions. The ensemble is shifted by distinct mutational events, cofactors, post-translational modifications, and different membrane compositions. A better understanding of Ras biology can contribute to therapeutic strategies.
Collapse
Affiliation(s)
- Shaoyong Lu
- Department of Pathophysiology, Shanghai Universities E-Institute for Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine , Shanghai, 200025, China.,Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, National Cancer Institute , Frederick, Maryland 21702, United States
| | - Hyunbum Jang
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, National Cancer Institute , Frederick, Maryland 21702, United States
| | | | | | | | - Ruth Nussinov
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, National Cancer Institute , Frederick, Maryland 21702, United States.,Department of Human Genetics and Molecular Medicine, Sackler School of Medicine, Sackler Institute of Molecular Medicine, Tel Aviv University , Tel Aviv 69978, Israel
| | - Jian Zhang
- Department of Pathophysiology, Shanghai Universities E-Institute for Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine , Shanghai, 200025, China
| |
Collapse
|
35
|
Strucken EM, Lee SH, Jang GW, Porto-Neto LR, Gondro C. Towards breed formation by island model divergence in Korean cattle. BMC Evol Biol 2015; 15:284. [PMID: 26677975 PMCID: PMC4683938 DOI: 10.1186/s12862-015-0563-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/08/2015] [Indexed: 01/03/2023] Open
Abstract
Background The main cattle breed in Korea is the brown Hanwoo, which has been under artificial selection within a national breeding program for several decades. Varieties of the Hanwoo known as Jeju Black and Chikso were not included in the breeding program and remained isolated from the effects of recent artificial selection advancements. We analysed the Jeju Black and Chikso populations in regards to their genetic variability, state of inbreeding, as well as level of differentiation from the mainland Hanwoo population. Results Jeju Black and Chikso were found to have small estimated effective population sizes (Ne) of only 11 and 7, respectively. Despite a small Ne, higher than expected heterozygosity levels were observed (0.303 and 0.306), however, lower allelic richness was found for the two island populations (1.76 and 1.77) compared to the mainland population (1.81). The increase in heterozygosity could be due to environmental disease challenges that promoted maintenance of higher genetic variability; however, no direct proof exists. Increased heterozygosity due to a first generation crossing of genetically different populations is not recorded. The differentiation between the Korean populations had FST values between 0.014 and 0.036 which is not as high as the differentiation within European beef or dairy cattle breeds (0.047–0.111). This suggests that the three populations have not separated into independent breeds. Conclusion Results agree with an island model of speciation where the brown Hanwoo represents the ancestral breed, whilst the Jeju Black and Chikso diverge from this common ancestor, following different evolutionary trajectories. Nevertheless, differences are minor and whether Jeju Black and Chikso cattle will develop into discrete breeds or reintegrate with the main population has to be seen in the future and will largely depend on human management decisions. This offers a rare opportunity to accompany the development of new breeds but also poses challenges on how to preserve these incipient breeds and ensure their long term viability. Electronic supplementary material The online version of this article (doi:10.1186/s12862-015-0563-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Eva M Strucken
- University of New England, School of Environmental and Rural Science, Armidale, Australia.
| | - Seung H Lee
- Division of Animal & Dairy Science, Chungnam National University, Daejeon, Korea.
| | - Gul W Jang
- Rural Development Administration, National Institute of Animal Science, Wanju, Korea.
| | - Laercio R Porto-Neto
- Scientific and Industrial Research Organization, Agricultural Flagship, Brisbane, Australia.
| | - Cedric Gondro
- University of New England, School of Environmental and Rural Science, Armidale, Australia.
| |
Collapse
|
36
|
Genetics of non-conventional lipoprotein fractions. CURRENT GENETIC MEDICINE REPORTS 2015; 3:196-201. [PMID: 26618077 DOI: 10.1007/s40142-015-0077-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Lipoprotein subclass measures associate with cardiometabolic disease risk. Currently the information that lipoproteins convey on disease risk over that of traditional demographic and lipid measures is minimal, and so their use is clinics is limited. However, lipoprotein subclass perturbations represent some of the earliest manifestations of metabolic dysfunction, and their etiology is partially distinct from lipids, so information on the genetic etiology of lipoproteins offers promise for improved risk prediction, and unique mechanistic insights into IR and atherosclerosis. Here, I review the genetic variants validated as associating with lipoprotein measures to date, and show that the majority of identified variants have functionality that is best understood as related to lipid measures. Until we focus on the genes as they relate to lipoprotein subclass production, we are limiting our understanding of biological mechanisms underlying cardiometabolic disease.
Collapse
|
37
|
Watson GM, Gunzburg MJ, Ambaye ND, Lucas WAH, Traore DA, Kulkarni K, Cergol KM, Payne RJ, Panjikar S, Pero SC, Perlmutter P, Wilce MCJ, Wilce JA. Cyclic Peptides Incorporating Phosphotyrosine Mimetics as Potent and Specific Inhibitors of the Grb7 Breast Cancer Target. J Med Chem 2015; 58:7707-18. [DOI: 10.1021/acs.jmedchem.5b00609] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
| | | | | | | | | | | | - Katie M. Cergol
- School
of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Richard J. Payne
- School
of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Santosh Panjikar
- Australian Synchrotron, 800 Blackburn
Road, Clayton, Victoria 3168, Australia
| | - Stephanie C. Pero
- Department
of Surgery and Vermont Cancer Center, University of Vermont, Burlington, Vermont 05401, United States
| | | | | | | |
Collapse
|
38
|
Seim I, Ma S, Zhou X, Gerashchenko MV, Lee SG, Suydam R, George JC, Bickham JW, Gladyshev VN. The transcriptome of the bowhead whale Balaena mysticetus reveals adaptations of the longest-lived mammal. Aging (Albany NY) 2015; 6:879-99. [PMID: 25411232 PMCID: PMC4247388 DOI: 10.18632/aging.100699] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mammals vary dramatically in lifespan, by at least two-orders of magnitude, but the molecular basis for this difference remains largely unknown. The bowhead whale Balaena mysticetus is the longest-lived mammal known, with an estimated maximal lifespan in excess of two hundred years. It is also one of the two largest animals and the most cold-adapted baleen whale species. Here, we report the first genome-wide gene expression analyses of the bowhead whale, based on the de novo assembly of its transcriptome. Bowhead whale or cetacean-specific changes in gene expression were identified in the liver, kidney and heart, and complemented with analyses of positively selected genes. Changes associated with altered insulin signaling and other gene expression patterns could help explain the remarkable longevity of bowhead whales as well as their adaptation to a lipid-rich diet. The data also reveal parallels in candidate longevity adaptations of the bowhead whale, naked mole rat and Brandt's bat. The bowhead whale transcriptome is a valuable resource for the study of this remarkable animal, including the evolution of longevity and its important correlates such as resistance to cancer and other diseases.
Collapse
Affiliation(s)
- Inge Seim
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Siming Ma
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xuming Zhou
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Maxim V Gerashchenko
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sang-Goo Lee
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Robert Suydam
- Department of Wildlife Management, North Slope Borough, Barrow, AK 99723, USA
| | - John C George
- Department of Wildlife Management, North Slope Borough, Barrow, AK 99723, USA
| | | | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
39
|
Perdereau D, Cailliau K, Browaeys-Poly E, Lescuyer A, Carré N, Benhamed F, Goenaga D, Burnol AF. Insulin-induced cell division is controlled by the adaptor Grb14 in a Chfr-dependent manner. Cell Signal 2015; 27:798-806. [PMID: 25578860 DOI: 10.1016/j.cellsig.2015.01.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 01/03/2015] [Indexed: 01/02/2023]
Abstract
Beyond its key role in the control of energy metabolism, insulin is also an important regulator of cell division and neoplasia. However, the molecular events involved in insulin-driven cell proliferation are not fully elucidated. Here, we show that the ubiquitin ligase Chfr, a checkpoint protein involved in G2/M transition, is a new effector involved in the control of insulin-induced cell proliferation. Chfr is identified as a partner of the molecular adapter Grb14, an inhibitor of insulin signalling. Using mammalian cell lines and the Xenopus oocyte as a model of G2/M transition, we demonstrate that Chfr potentiates the inhibitory effect of Grb14 on insulin-induced cell division. Insulin stimulates Chfr binding to the T220 residue of Grb14. Both Chfr binding site and Grb14 C-ter BPS-SH2 domain, mediating IR binding and inhibition, are required to prevent insulin-induced cell division. Targeted mutagenesis revealed that Chfr ligase activity and phosphorylation of its T39 residue, a target of Akt, are required to potentiate Grb14 inhibitory activity. In the presence of insulin, the binding of Chfr to Grb14 activates its ligase activity, leading to Aurora A and Polo-like kinase degradation and blocking cell division. Collectively, our results show that Chfr and Grb14 collaborate in a negative feedback loop controlling insulin-stimulated cell division.
Collapse
Affiliation(s)
- Dominique Perdereau
- INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité; 24, Rue du Faubourg Saint Jacques, Paris 75014, France
| | - Katia Cailliau
- Laboratoire de Régulation des Signaux de Division, Université de Lille 1, UE 4479, IFR 147, Villeneuve d'Ascq 59655, France
| | - Edith Browaeys-Poly
- Laboratoire de Régulation des Signaux de Division, Université de Lille 1, UE 4479, IFR 147, Villeneuve d'Ascq 59655, France
| | - Arlette Lescuyer
- Laboratoire de Régulation des Signaux de Division, Université de Lille 1, UE 4479, IFR 147, Villeneuve d'Ascq 59655, France
| | - Nadège Carré
- INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité; 24, Rue du Faubourg Saint Jacques, Paris 75014, France
| | - Fadila Benhamed
- INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité; 24, Rue du Faubourg Saint Jacques, Paris 75014, France
| | - Diana Goenaga
- INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité; 24, Rue du Faubourg Saint Jacques, Paris 75014, France
| | - Anne-Françoise Burnol
- INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité; 24, Rue du Faubourg Saint Jacques, Paris 75014, France.
| |
Collapse
|
40
|
Ali O, Cerjak D, Kent JW, James R, Blangero J, Carless MA, Zhang Y. An epigenetic map of age-associated autosomal loci in northern European families at high risk for the metabolic syndrome. Clin Epigenetics 2015; 7:12. [PMID: 25806089 PMCID: PMC4372177 DOI: 10.1186/s13148-015-0048-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 01/16/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The prevalence of chronic diseases such as cancer, type 2 diabetes, metabolic syndrome (MetS), and cardiovascular disease increases with age in all populations. Epigenetic features are hypothesized to play important roles in the pathophysiology of age-associated diseases, but a map of these markers is lacking. We searched for genome-wide age-associated methylation signatures in peripheral blood of individuals at high risks for MetS by profiling 485,000 CpG sites in 192 individuals of Northern European ancestry using the Illumina HM450 array. Subjects (ages 6-85 years) were part of seven extended families, and 73% of adults and 32% of children were overweight or obese. RESULTS We found 22,122 genome-wide significant age-associated CpG sites (P α=0.05 = 3.65 × 10(-7) after correction for multiple testing) of which 14,155 are positively associated with age while 7,967 are negatively associated. By applying a positional density-based clustering algorithm, we generated a map of epigenetic 'hot-spots' of age-associated genomic segments, which include 290 age-associated differentially methylated CpG clusters (aDMCs), of which 207 are positively associated with age. Gene/pathway enrichment analyses were performed on these clusters using FatiGO. Genes localized to both the positively (n = 241) and negatively (n = 16) age-associated clusters are significantly enriched in specific KEGG pathways and GO terms. The most significantly enriched pathways are the hedgehog signaling pathway (adjusted P = 3.96 × 10(-3)) and maturity-onset diabetes of the young (MODY) (adjusted P = 6.26 × 10(-3)) in the positive aDMCs and type I diabetes mellitus (adjusted P = 3.69 × 10(-7)) in the negative aDMCs. We also identified several epigenetic loci whose age-associated change rates differ between subjects diagnosed with MetS and those without. CONCLUSION We conclude that in a family cohort at high risk for MetS, age-associated epigenetic features enrich in biological pathways important for determining the fate of fat cells and for insulin production. We also observe that several genes known to be related to MetS show differential epigenetic response to age in individuals with and without MetS.
Collapse
Affiliation(s)
- Omar Ali
- />Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin USA
| | - Diana Cerjak
- />TOPS Obesity and Metabolic Research Center, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin USA
- />Human and Molecular Genetics Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, Wisconsin USA
| | - Jack W Kent
- />Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas USA
| | - Roland James
- />TOPS Obesity and Metabolic Research Center, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin USA
- />Human and Molecular Genetics Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, Wisconsin USA
| | - John Blangero
- />Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas USA
| | - Melanie A Carless
- />Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas USA
| | - Yi Zhang
- />TOPS Obesity and Metabolic Research Center, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin USA
- />Human and Molecular Genetics Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, Wisconsin USA
| |
Collapse
|
41
|
Effect of knocking down the insulin receptor on mouse rod responses. Sci Rep 2015; 5:7858. [PMID: 25598343 PMCID: PMC4297982 DOI: 10.1038/srep07858] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 12/18/2014] [Indexed: 12/12/2022] Open
Abstract
Previous experiments have shown that the insulin receptor (IR) is expressed in mammalian rods and contributes to the protection of photoreceptors during bright-light exposure. The role of the insulin receptor in the production of the light response is however unknown. We have used suction-electrode recording to examine the responses of rods after conditionally knocking down the insulin receptor. Our results show that these IR knock-down rods have an accelerated decay of the light response and a small decrease in sensitivity by comparison to littermate WT rods. Our results indicate that the insulin receptor may have some role in controlling the rate of rod response decay, but they exclude a major role of the insulin receptor pathway in phototransduction.
Collapse
|
42
|
Kalaivani R, Srinivasan N. A Gaussian network model study suggests that structural fluctuations are higher for inactive states than active states of protein kinases. MOLECULAR BIOSYSTEMS 2015; 11:1079-95. [DOI: 10.1039/c4mb00675e] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Protein kinases participate extensively in cellular signalling. Using Gaussian normal mode analysis of kinases in active and diverse inactive forms, authors show that structural fluctuations are significantly higher in inactive forms and are localized in functionally sensitive sites.
Collapse
Affiliation(s)
- Raju Kalaivani
- Molecular Biophysics Unit
- Indian Institute of Science
- Bangalore 560012
- India
| | | |
Collapse
|
43
|
Kavran JM, McCabe JM, Byrne PO, Connacher MK, Wang Z, Ramek A, Sarabipour S, Shan Y, Shaw DE, Hristova K, Cole PA, Leahy DJ. How IGF-1 activates its receptor. eLife 2014; 3:03772. [PMID: 25255214 PMCID: PMC4381924 DOI: 10.7554/elife.03772] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 09/23/2014] [Indexed: 12/20/2022] Open
Abstract
The type I insulin-like growth factor receptor (IGF1R) is involved in growth and survival of normal and neoplastic cells. A ligand-dependent conformational change is thought to regulate IGF1R activity, but the nature of this change is unclear. We point out an underappreciated dimer in the crystal structure of the related Insulin Receptor (IR) with Insulin bound that allows direct comparison with unliganded IR and suggests a mechanism by which ligand regulates IR/IGF1R activity. We test this mechanism in a series of biochemical and biophysical assays and find the IGF1R ectodomain maintains an autoinhibited state in which the TMs are held apart. Ligand binding releases this constraint, allowing TM association and unleashing an intrinsic propensity of the intracellular regions to autophosphorylate. Enzymatic studies of full-length and kinase-containing fragments show phosphorylated IGF1R is fully active independent of ligand and the extracellular-TM regions. The key step triggered by ligand binding is thus autophosphorylation.
Collapse
Affiliation(s)
- Jennifer M Kavran
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Jacqueline M McCabe
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Patrick O Byrne
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Mary Katherine Connacher
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Zhihong Wang
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, United States.,Department of Chemistry and Biochemistry, University of the Sciences, Philadelphia, United States
| | | | - Sarvenaz Sarabipour
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, United States
| | | | - David E Shaw
- DE Shaw Research, New York, United States.,Department of Biochemistry and Molecular Biophysics, Columbia University, New York, United States
| | - Kalina Hristova
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, United States
| | - Philip A Cole
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Daniel J Leahy
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, United States.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, United States
| |
Collapse
|
44
|
Gondoin A, Morzyglod L, Desbuquois B, Burnol AF. [Control of insulin signalisation and action by the Grb14 protein]. Biol Aujourdhui 2014; 208:119-36. [PMID: 25190572 DOI: 10.1051/jbio/2014013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Indexed: 11/15/2022]
Abstract
The action of insulin on metabolism and cell growth is mediated by a specific receptor tyrosine kinase, which, through phosphorylation of several substrates, triggers the activation of two major signaling pathways, the phosphatidylinositol 3-kinase (PI3-K)/Akt pathway and the Ras/extracellular signal-regulated kinase (ERK) pathway. Insulin-induced activation of the receptor and downstream signaling is also subjected to a negative feedback control involving several mechanisms, among which the interaction of the insulin receptor and its substrates with inhibitory proteins. After summarizing the major mechanisms underlying the activation and attenuation of insulin signaling, this review focuses on its control by the Grb14 adaptor protein. Grb14 has been identif-ied as an inhibitor of insulin signaling and action, and is involved in insulin resistance associated with type 2 diabetes and obesity. Studies on the molecular mechanism of action of Grb14 have shown that, through interaction with the activated insulin receptor, Grb14 inhibits its catalytic activity and the activation of downstream signaling. However, the consequences of Grb14 gene invalidation are complex and tissue-specific, and some effects of Grb14 on insulin signaling appear to be linked to its interaction with effector proteins downstream the insulin receptor. Pharmacological inhibition of Grb14 should allow to enhance insulin sensitivity and improve energy homeostasis in insulin-resistant states.
Collapse
Affiliation(s)
- Anaïs Gondoin
- INSERM, U1016, Institut Cochin, 22 rue Méchain, 75014 Paris, France - CNRS, UMR 8104, Institut Cochin, 22 rue Méchain, 75014 Paris, France - Université Paris Descartes, Sorbonne Paris Cité, 24 rue du Faubourg Saint Jacques, 75014 Paris, France
| | - Lucie Morzyglod
- INSERM, U1016, Institut Cochin, 22 rue Méchain, 75014 Paris, France - CNRS, UMR 8104, Institut Cochin, 22 rue Méchain, 75014 Paris, France - Université Paris Descartes, Sorbonne Paris Cité, 24 rue du Faubourg Saint Jacques, 75014 Paris, France
| | - Bernard Desbuquois
- INSERM, U1016, Institut Cochin, 22 rue Méchain, 75014 Paris, France - CNRS, UMR 8104, Institut Cochin, 22 rue Méchain, 75014 Paris, France - Université Paris Descartes, Sorbonne Paris Cité, 24 rue du Faubourg Saint Jacques, 75014 Paris, France
| | - Anne-Françoise Burnol
- INSERM, U1016, Institut Cochin, 22 rue Méchain, 75014 Paris, France - CNRS, UMR 8104, Institut Cochin, 22 rue Méchain, 75014 Paris, France - Université Paris Descartes, Sorbonne Paris Cité, 24 rue du Faubourg Saint Jacques, 75014 Paris, France
| |
Collapse
|
45
|
Kraja AT, Chasman DI, North KE, Reiner AP, Yanek LR, Kilpeläinen TO, Smith JA, Dehghan A, Dupuis J, Johnson AD, Feitosa MF, Tekola-Ayele F, Chu AY, Nolte IM, Dastani Z, Morris A, Pendergrass SA, Sun YV, Ritchie MD, Vaez A, Lin H, Ligthart S, Marullo L, Rohde R, Shao Y, Ziegler MA, Im HK, Schnabel RB, Jørgensen T, Jørgensen ME, Hansen T, Pedersen O, Stolk RP, Snieder H, Hofman A, Uitterlinden AG, Franco OH, Ikram MA, Richards JB, Rotimi C, Wilson JG, Lange L, Ganesh SK, Nalls M, Rasmussen-Torvik LJ, Pankow JS, Coresh J, Tang W, Linda Kao WH, Boerwinkle E, Morrison AC, Ridker PM, Becker DM, Rotter JI, Kardia SLR, Loos RJF, Larson MG, Hsu YH, Province MA, Tracy R, Voight BF, Vaidya D, O'Donnell CJ, Benjamin EJ, Alizadeh BZ, Prokopenko I, Meigs JB, Borecki IB. Pleiotropic genes for metabolic syndrome and inflammation. Mol Genet Metab 2014; 112:317-38. [PMID: 24981077 PMCID: PMC4122618 DOI: 10.1016/j.ymgme.2014.04.007] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 04/26/2014] [Accepted: 04/26/2014] [Indexed: 01/11/2023]
Abstract
Metabolic syndrome (MetS) has become a health and financial burden worldwide. The MetS definition captures clustering of risk factors that predict higher risk for diabetes mellitus and cardiovascular disease. Our study hypothesis is that additional to genes influencing individual MetS risk factors, genetic variants exist that influence MetS and inflammatory markers forming a predisposing MetS genetic network. To test this hypothesis a staged approach was undertaken. (a) We analyzed 17 metabolic and inflammatory traits in more than 85,500 participants from 14 large epidemiological studies within the Cross Consortia Pleiotropy Group. Individuals classified with MetS (NCEP definition), versus those without, showed on average significantly different levels for most inflammatory markers studied. (b) Paired average correlations between 8 metabolic traits and 9 inflammatory markers from the same studies as above, estimated with two methods, and factor analyses on large simulated data, helped in identifying 8 combinations of traits for follow-up in meta-analyses, out of 130,305 possible combinations between metabolic traits and inflammatory markers studied. (c) We performed correlated meta-analyses for 8 metabolic traits and 6 inflammatory markers by using existing GWAS published genetic summary results, with about 2.5 million SNPs from twelve predominantly largest GWAS consortia. These analyses yielded 130 unique SNPs/genes with pleiotropic associations (a SNP/gene associating at least one metabolic trait and one inflammatory marker). Of them twenty-five variants (seven loci newly reported) are proposed as MetS candidates. They map to genes MACF1, KIAA0754, GCKR, GRB14, COBLL1, LOC646736-IRS1, SLC39A8, NELFE, SKIV2L, STK19, TFAP2B, BAZ1B, BCL7B, TBL2, MLXIPL, LPL, TRIB1, ATXN2, HECTD4, PTPN11, ZNF664, PDXDC1, FTO, MC4R and TOMM40. Based on large data evidence, we conclude that inflammation is a feature of MetS and several gene variants show pleiotropic genetic associations across phenotypes and might explain a part of MetS correlated genetic architecture. These findings warrant further functional investigation.
Collapse
Affiliation(s)
- Aldi T Kraja
- Division of Statistical Genomics, Department of Genetics and Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA.
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Kari E North
- Department of Epidemiology and Carolina Center for Genome Sciences, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, NC, USA.
| | | | - Lisa R Yanek
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Tuomas O Kilpeläinen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Jennifer A Smith
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA.
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA; National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA.
| | - Andrew D Johnson
- National Heart, Lung and Blood Institute (NHLBI) Division of Intramural Research and NHLBI's Framingham Heart Study, Framingham, MA, USA.
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics and Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA.
| | - Fasil Tekola-Ayele
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Audrey Y Chu
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Ilja M Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Zari Dastani
- Department of Epidemiology, Biostatistics and Occupational Health, Jewish General Hospital, Lady Davis Institute, McGill University Montreal, Quebec, Canada.
| | - Andrew Morris
- The Welcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK.
| | - Sarah A Pendergrass
- Department of Biochemistry and Molecular Biology, Eberly College of Science and The Huck Institutes of the Life Sciences, The Pennsylvania State University, PA, USA.
| | - Yan V Sun
- Department of Epidemiology, Rollins School of Public Health, and Department of Biomedical Informatics, School of Medicine, Emory University, Atlanta, GA, USA.
| | - Marylyn D Ritchie
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, USA.
| | - Ahmad Vaez
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Honghuang Lin
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA.
| | - Symen Ligthart
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Letizia Marullo
- The Welcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK; Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy.
| | - Rebecca Rohde
- Department of Epidemiology and Carolina Center for Genome Sciences, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, NC, USA.
| | - Yaming Shao
- Department of Epidemiology and Carolina Center for Genome Sciences, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, NC, USA.
| | - Mark A Ziegler
- Division of Biostatistics, MSIBS Program, Washington University School of Medicine, St. Louis, MO, USA.
| | - Hae Kyung Im
- Department of Health Studies, University of Chicago, IL, USA.
| | - Renate B Schnabel
- Department of General and Interventional Cardiology University Heart Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Torben Jørgensen
- Research Centre for Prevention and Health, Glostrup Hospital, Glostrup, Denmark; Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark.
| | | | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Oluf Pedersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Ronald P Stolk
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Andre G Uitterlinden
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Oscar H Franco
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - J Brent Richards
- Department of Epidemiology, Biostatistics and Occupational Health, Jewish General Hospital, Lady Davis Institute, McGill University Montreal, Quebec, Canada; Department of Medicine, Human Genetics, Epidemiology and Biostatistics, McGill University, Canada; Department of Twin Research, King's College, London, UK.
| | - Charles Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| | | | - Leslie Lange
- Department of Genetics, University of North Carolina, NC, USA.
| | - Santhi K Ganesh
- Department of Internal Medicine, University of Michigan, MI, USA.
| | - Mike Nalls
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD, USA.
| | | | - James S Pankow
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, USA.
| | - Josef Coresh
- Department of Medicine, Epidemiology, Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA.
| | - Weihong Tang
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, USA.
| | - W H Linda Kao
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | - Eric Boerwinkle
- Human Genetics Center, University of Texas - Houston Health Science Center at Houston, Houston, TX, USA.
| | - Alanna C Morrison
- Human Genetics Center, University of Texas - Houston Health Science Center at Houston, Houston, TX, USA.
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Diane M Becker
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute (LA BioMed), Harbor-UCLA Medical Center, Torrance, CA, USA.
| | - Sharon L R Kardia
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA.
| | - Ruth J F Loos
- The Genetics of Obesity and Related Metabolic Traits Program, The Charles Bronfman Institute for Personalized Medicine, The Mindich Child Health and Development Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Martin G Larson
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA; National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; Department of Mathematics and Statistics, Boston University, Boston, MA, USA.
| | - Yi-Hsiang Hsu
- Hebrew Senior Life Institute for Aging Research, Harvard Medical School and Molecular and Integrative Physiological Sciences, Harvard School of Public Health, Boston, MA, USA.
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics and Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA.
| | - Russell Tracy
- University of Vermont College of Medicine, Burlington, VT, USA.
| | - Benjamin F Voight
- Department of Pharmacology, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, USA; Department of Genetics, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, USA.
| | - Dhananjay Vaidya
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Christopher J O'Donnell
- National Heart, Lung and Blood Institute (NHLBI) Division of Intramural Research and NHLBI's Framingham Heart Study, Framingham, MA, USA.
| | - Emelia J Benjamin
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; Cardiology and Preventive Medicine Sections, Department of Medicine, Boston University School of Medicine, Boston, MA, USA.
| | - Behrooz Z Alizadeh
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Inga Prokopenko
- Department of Genomics of Common Diseases, School of Public Health, Imperial College London, London W12 0NN, UK.
| | - James B Meigs
- General Medicine Division, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Ingrid B Borecki
- Division of Statistical Genomics, Department of Genetics and Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
46
|
Mukherjee M, Jing-Song F, Ramachandran S, Guy GR, Sivaraman J. Dimeric switch of Hakai-truncated monomers during substrate recognition: insights from solution studies and NMR structure. J Biol Chem 2014; 289:25611-23. [PMID: 25074933 DOI: 10.1074/jbc.m114.592840] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hakai, an E3 ubiquitin ligase, disrupts cell-cell contacts in epithelial cells and is up-regulated in human colon and gastric adenocarcinomas. Hakai acts through its phosphotyrosine-binding (HYB) domain, which bears a dimeric fold that recognizes the phosphotyrosine motifs of E-cadherin, cortactin, DOK1, and other Src substrates. Unlike the monomeric nature of the SH2 and phosphotyrosine-binding domains, the architecture of the HYB domain consists of an atypical, zinc-coordinated tight homodimer. Here, we report a C-terminal truncation mutant of the HYB domain (HYB(ΔC)), comprising amino acids 106-194, which exists as a monomer in solution. The NMR structure revealed that this deletion mutant undergoes a dramatic structural change caused by a rearrangement of the atypical zinc-coordinated unit in the C terminus of the HYB domain to a C2H2-like zinc finger in HYB(ΔC). Moreover, using isothermal titration calorimetry, we show that dimerization of HYB(ΔC) can be induced using a phosphotyrosine substrate peptide. This ligand-induced dimerization of HYB(ΔC) is further validated using analytical ultracentrifugation, size-exclusion chromatography, NMR relaxation studies, dynamic light scattering, and circular dichroism experiments. Overall, these observations suggest that the dimeric architecture of the HYB domain is essential for the phosphotyrosine-binding property of Hakai.
Collapse
Affiliation(s)
- Manjeet Mukherjee
- From the Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore 117543 and
| | - Fan Jing-Song
- From the Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore 117543 and
| | - Sarath Ramachandran
- From the Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore 117543 and
| | - Graeme R Guy
- the Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Singapore 138673
| | - J Sivaraman
- From the Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore 117543 and
| |
Collapse
|
47
|
Taira J, Higashimoto Y. Phosphorylation of Grb14 BPS domain by GSK-3 correlates with complex forming of Grb14 and insulin receptor. J Biochem 2014; 155:353-60. [PMID: 24535599 DOI: 10.1093/jb/mvu011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Growth factor receptor-bound protein 14 (Grb14) interacts with insulin receptor (IR) through the between PH and SH2 (BPS) domain. Grb14-IR complex formation is initiated by insulin stimulation, and the binding event results in the inhibition of insulin signalling. Thus, Grb14 is regarded as an endogenous suppressor of insulin signal transduction; however, there are no studies describing the mechanism whereby Grb14-IR complex formation is suppressed in the absence of insulin stimulation. In the present study, multiple phosphorylation motifs for glycogen synthase kinase 3 (GSK-3) were identified within the Grb14 BPS domain (Ser(358), Ser(362) and Ser(366) of human Grb14). Pharmacological inhibition as well as knockdown of GSK-3 facilitated complex formation between Grb14 and IR, implicating GSK-3 activity in regulating Grb14-IR binding. In situ proximity ligation assay and in vitro kinase assays of phosphopeptides suggested that serine residues in the BPS domain would be substrates for GSK-3. The kinase assays also indicated phosphoserine 370 (in human Grb14) was required for the phosphorylation of Ser(358), Ser(362) and Ser(366) by GSK-3. Grb14-IR binding was also facilitated by replacement of the serines with Ala. We also observed that Ser(366) of endogenous Grb14 in Hep G2 cell was phosphorylated and the phosphorylation was influenced by treatments with insulin, as well as the GSK-3 inhibitor.
Collapse
Affiliation(s)
- Junichi Taira
- Department of Chemistry, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan
| | - Yuichiro Higashimoto
- Department of Chemistry, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan
| |
Collapse
|
48
|
Babon JJ, Varghese LN, Nicola NA. Inhibition of IL-6 family cytokines by SOCS3. Semin Immunol 2014; 26:13-9. [PMID: 24418198 DOI: 10.1016/j.smim.2013.12.004] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 12/23/2013] [Indexed: 01/06/2023]
Abstract
IL-6 a multi-functional cytokine with important effects in both inflammation and haematopoiesis. SOCS3 is the primary inhibitor of IL-6 signalling, interacting with gp130, the common shared chain of the IL-6 family of cytokines, and JAK1, JAK2 and TYK2 to control both the duration of signalling and the biological response. Recent biochemical and structural studies have shown SOCS3 binds to only these three JAKs, all of which are associated with IL-6 signalling, and not JAK3. This specificity is determined by a three residue "GQM" motif in the kinase domain of JAK1, JAK2 and TYK2. SOCS3 binds to JAK and gp130 simultaneously, and inhibits JAK activity in an ATP-independent manner by partially occluding the kinase's substrate binding groove with its kinase inhibitory region. We therefore propose a model in which each of gp130, JAK and SOCS3 are directly bound to the other two, allowing SOCS3 to inhibit IL6 signalling with high potency and specificity.
Collapse
Affiliation(s)
- Jeffrey J Babon
- Walter and Eliza Hall Institute, Parkville, Australia; The University of Melbourne, Parkville, Australia.
| | - Leila N Varghese
- Walter and Eliza Hall Institute, Parkville, Australia; The University of Melbourne, Parkville, Australia
| | - Nicos A Nicola
- Walter and Eliza Hall Institute, Parkville, Australia; The University of Melbourne, Parkville, Australia
| |
Collapse
|
49
|
Rajala RVS, Basavarajappa DK, Dighe R, Rajala A. Spatial and temporal aspects and the interplay of Grb14 and protein tyrosine phosphatase-1B on the insulin receptor phosphorylation. Cell Commun Signal 2013; 11:96. [PMID: 24350791 PMCID: PMC3878334 DOI: 10.1186/1478-811x-11-96] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 12/15/2013] [Indexed: 12/17/2022] Open
Abstract
Background Growth factor receptor-bound protein 14 (Grb14) is an adapter protein implicated in receptor tyrosine kinase signaling. Grb14 knockout studies highlight both the positive and negative roles of Grb14 in receptor tyrosine kinase signaling, in a tissue specific manner. Retinal cells are post-mitotic tissue, and insulin receptor (IR) activation is essential for retinal neuron survival. Retinal cells express protein tyrosine phosphatase-1B (PTP1B), which dephosphorylates IR and Grb14, a pseudosubstrate inhibitor of IR. This project asks the following major question: in retinal neurons, how does the IR overcome inactivation by PTP1B and Grb14? Results Our previous studies suggest that ablation of Grb14 results in decreased IR activation, due to increased PTP1B activity. Our research propounds that phosphorylation in the BPS region of Grb14 inhibits PTP1B activity, thereby promoting IR activation. We propose a model in which phosphorylation of the BPS region of Grb14 is the key element in promoting IR activation, and failure to undergo phosphorylation on Grb14 leads to both PTP1B and Grb14 exerting their negative roles in IR. Consistent with this hypothesis, we found decreased phosphorylation of Grb14 in diabetic type 1 Ins2Akita mouse retinas. Decreased retinal IR activation has previously been reported in this mouse line. Conclusions Our results suggest that phosphorylation status of the BPS region of Grb14 determines the positive or negative role it will play in IR signaling.
Collapse
Affiliation(s)
- Raju V S Rajala
- Departments of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| | | | | | | |
Collapse
|
50
|
Huang O, Jiang M, Zhang X, Xie Z, Chen X, Wu J, Liu H, Shen K. Grb14 as an independent good prognosis factor for breast cancer patients treated with neoadjuvant chemotherapy. Jpn J Clin Oncol 2013; 43:1064-72. [PMID: 24031083 DOI: 10.1093/jjco/hyt130] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVE Growth factor receptor-binding protein 14, a new member of noncatalytic adaptor proteins family, has been shown to be upregulated in breast cancer. We investigated the prognostic value of growth factor receptor-binding protein 14 expression in breast cancer patients treated with neoadjuvant chemotherapy. METHODS Primary breast cancer specimens were taken from locally advanced breast cancer patients in a Phase II clinical trial of neoadjuvant chemotherapy and the expression pattern of growth factor receptor-binding protein 14 was determined by immunohistochemistry. Kaplan-Meier analysis and Cox regression model were used to assess disease-free and overall survival, according to the expression of growth factor receptor-binding protein 14 in tumor cells. RESULTS Our result showed that growth factor receptor-binding protein 14 was highly expressed in 23.1% of breast cancer sections, and high expression of growth factor receptor-binding protein 14 was significantly associated with better disease-free (P = 0.016, hazard ratio 0.07, 95% confidence interval 0.06-0.08) and overall survival (P = 0.004, hazard ratio 0.02, 95% confidence interval 0.02-0.03), compared with the low-expression group. Multivariate analysis indicated that high expression of growth factor receptor-binding protein 14 was an independent good prognostic factor for both disease-free (P = 0.04, hazard ratio 0.37, 95% confidence interval 0.14-0.98) and overall survival (P = 0.03, hazard ratio 0.11, 95% confidence interval 0.10-0.82). CONCLUSIONS High expression of growth factor receptor-binding protein 14 in breast cancer cells may help to identify low-risk patients for additional therapies after neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Ou Huang
- *Department of Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, No.197, 2nd Ruijin Road, Huangpu District, Shanghai 200025, PR China.
| | | | | | | | | | | | | | | |
Collapse
|