1
|
Zhai T, Cai L, Jia X, Xia M, Bian H, Gao X, Pan C, Li X, Xia P. IGFBP2 functions as an endogenous protector against hepatic steatosis via suppression of the EGFR-STAT3 pathway. Mol Metab 2024; 89:102026. [PMID: 39299533 PMCID: PMC11474195 DOI: 10.1016/j.molmet.2024.102026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/17/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024] Open
Abstract
OBJECTIVE Non-alcoholic fatty liver disease (NAFLD) is deemed as an emerging global epidemic, whereas the underlying pathogenic mechanism remains to be clarified. We aimed to systemically analyze all the NAFLD-related gene expression datasets from published human-based studies, by which exploring potential key factors and mechanisms accounting for the pathogenesis of NAFLD. METHODS Robust rank aggregation (RRA) method was used to integrate NAFLD-related gene expression datasets. For fatty liver study, adeno-associated virus (AAV) delivery and genetic knockout mice were used to create IGFBP2 (Insulin-like growth factor binding protein 2) gain- or loss-of function models. Western blot, Co-immunoprecipitation (Co-IP), immunofluorescent (IF) staining, luciferase assay, molecular docking simulation were performed to reveal the IGFBP2-EGFR-STAT3 axis involved. Key axis protein levels in livers from healthy donors and patients with NAFLD were assessed via immunohistochemical staining. RESULTS By using RRA method, the present study identified IGFBP2 being the most significantly down-regulated gene in all NAFLD subjects. The decreased IGFBP2 expression was further confirmed in the liver tissues from patients and animal models of NAFLD. IGFBP2 deficiency aggravated hepatic steatosis and NASH phenotypes and promoted lipogenic gene expression both in vivo and in vitro. Mechanistically, IGFBP2 directly binds to and regulates EGFR, whereas blockage of the IGFBP2-EGFR complex by knockdown of IGFBP2 resulted in the EGFR-STAT3 pathway activation, which in turn promoted the promoter activity of Srebf1. By using molecular docking simulation and protein-protein interaction analysis, the sequence of 233-257 amino acids in IGFBP2 was characterized as a key motif responding for its specific binding to EGFR and the protective effect against hepatic steatosis. CONCLUSIONS The current study has, for the first time, identified IGFBP2 as a novel protector against hepatosteatosis. The protective effect is mediated by its specific interaction with EGFR and thereby suppressing the EGFR-STAT3 pathway. Therefore, pharmaceutically targeting the IGFBP2-EGFR-STAT3 axis may provide a theoretical basis for for the treatment of NAFLD/NASH and the associated diseases.
Collapse
Affiliation(s)
- Tianyu Zhai
- Department of Endocrinology and Metabolism, Zhongshan Hospital, and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China.
| | - Liang Cai
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Interventional Medicine (NCRC-IM), China; Institute of Vascular Surgery, Fudan University, Shanghai, China.
| | - Xi Jia
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong S.A.R, China.
| | - Mingfeng Xia
- Department of Endocrinology and Metabolism, Zhongshan Hospital, and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China.
| | - Hua Bian
- Department of Endocrinology and Metabolism, Zhongshan Hospital, and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China.
| | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital, and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China.
| | - Chenling Pan
- Beijing Computing Center, Beijing Academy of Science and Technology, Beijing, China.
| | - Xiaoying Li
- Department of Endocrinology and Metabolism, Zhongshan Hospital, and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China.
| | - Pu Xia
- Department of Endocrinology and Metabolism, Zhongshan Hospital, and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Rehman H, Ang TFA, Tao Q, Espenilla AL, Au R, Farrer LA, Zhang X, Qiu WQ. Comparison of Commonly Measured Plasma and Cerebrospinal Fluid Proteins and Their Significance for the Characterization of Cognitive Impairment Status. J Alzheimers Dis 2024; 97:621-633. [PMID: 38143358 DOI: 10.3233/jad-230837] [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] [Indexed: 12/26/2023]
Abstract
BACKGROUND Although cerebrospinal fluid (CSF) amyloid-β42 peptide (Aβ42) and phosphorylated tau (p-tau) and blood p-tau are valuable for differential diagnosis of Alzheimer's disease (AD) from cognitively normal (CN) there is a lack of validated biomarkers for mild cognitive impairment (MCI). OBJECTIVE This study sought to determine how plasma and CSF protein markers compared in the characterization of MCI and AD status. METHODS This cohort study included Alzheimer's Disease Neuroimaging Initiative (ADNI) participants who had baseline levels of 75 proteins measured commonly in plasma and CSF (257 total, 46 CN, 143 MCI, and 68 AD). Logistic regression, least absolute shrinkage and selection operator (LASSO) and Random Forest (RF) methods were used to identify the protein candidates for the disease classification. RESULTS We observed that six plasma proteins panel (APOE, AMBP, C3, IL16, IGFBP2, APOD) outperformed the seven CSF proteins panel (VEGFA, HGF, PRL, FABP3, FGF4, CD40, RETN) as well as AD markers (CSF p-tau and Aβ42) to distinguish the MCI from AD [area under the curve (AUC) = 0.75 (plasma proteins), AUC = 0.60 (CSF proteins) and AUC = 0.56 (CSF p-tau and Aβ42)]. Also, these six plasma proteins performed better than the CSF proteins and were in line with CSF p-tau and Aβ42 in differentiating CN versus MCI subjects [AUC = 0.89 (plasma proteins), AUC = 0.85 (CSF proteins) and AUC = 0.89 (CSF p-tau and Aβ42)]. These results were adjusted for age, sex, education, and APOEϵ4 genotype. CONCLUSIONS This study suggests that the combination of 6 plasma proteins can serve as an effective marker for differentiating MCI from AD and CN.
Collapse
Affiliation(s)
- Habbiburr Rehman
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ting Fang Alvin Ang
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Qiushan Tao
- Department of Pharmacology & Experimental Therapeutics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Arielle Lauren Espenilla
- Department of Biostatistics and Boston University School of Public Health, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Rhoda Au
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Framingham Heart Study, Boston University School of Medicine, Framingham, MA, USA
- Alzheimer's Disease Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Lindsay A Farrer
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics and Boston University School of Public Health, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Framingham Heart Study, Boston University School of Medicine, Framingham, MA, USA
- Alzheimer's Disease Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Xiaoling Zhang
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics and Boston University School of Public Health, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Framingham Heart Study, Boston University School of Medicine, Framingham, MA, USA
- Alzheimer's Disease Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Wei Qiao Qiu
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Pharmacology & Experimental Therapeutics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Alzheimer's Disease Research Center, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
3
|
Baxter RC. Signaling Pathways of the Insulin-like Growth Factor Binding Proteins. Endocr Rev 2023; 44:753-778. [PMID: 36974712 PMCID: PMC10502586 DOI: 10.1210/endrev/bnad008] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/25/2023] [Accepted: 03/15/2023] [Indexed: 03/29/2023]
Abstract
The 6 high-affinity insulin-like growth factor binding proteins (IGFBPs) are multifunctional proteins that modulate cell signaling through multiple pathways. Their canonical function at the cellular level is to impede access of insulin-like growth factor (IGF)-1 and IGF-2 to their principal receptor IGF1R, but IGFBPs can also inhibit, or sometimes enhance, IGF1R signaling either through their own post-translational modifications, such as phosphorylation or limited proteolysis, or by their interactions with other regulatory proteins. Beyond the regulation of IGF1R activity, IGFBPs have been shown to modulate cell survival, migration, metabolism, and other functions through mechanisms that do not appear to involve the IGF-IGF1R system. This is achieved by interacting directly or functionally with integrins, transforming growth factor β family receptors, and other cell-surface proteins as well as intracellular ligands that are intermediates in a wide range of pathways. Within the nucleus, IGFBPs can regulate the diverse range of functions of class II nuclear hormone receptors and have roles in both cell senescence and DNA damage repair by the nonhomologous end-joining pathway, thus potentially modifying the efficacy of certain cancer therapeutics. They also modulate some immune functions and may have a role in autoimmune conditions such as rheumatoid arthritis. IGFBPs have been proposed as attractive therapeutic targets, but their ubiquity in the circulation and at the cellular level raises many challenges. By understanding the diversity of regulatory pathways with which IGFBPs interact, there may still be therapeutic opportunities based on modulation of IGFBP-dependent signaling.
Collapse
Affiliation(s)
- Robert C Baxter
- Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital,St Leonards, NSW 2065, Australia
| |
Collapse
|
4
|
Nagasaki K, Gavrilova O, Hajishengallis G, Somerman MJ. Does the RGD region of certain proteins affect metabolic activity? FRONTIERS IN DENTAL MEDICINE 2022. [DOI: 10.3389/fdmed.2022.974862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A better understanding of the role of mineralized tissues and their associated factors in governing whole-body metabolism should be of value toward informing clinical strategies to treat mineralized tissue and metabolic disorders, such as diabetes and obesity. This perspective provides evidence suggesting a role for the arginine-glycine-aspartic acid (RGD) region, a sequence identified in several proteins secreted by bone cells, as well as other cells, in modulating systemic metabolic activity. We focus on (a) two of the SIBLING (small integrin-binding ligand, N-linked glycoprotein) family genes/proteins, bone sialoprotein (BSP) and osteopontin (OPN), (b) insulin-like growth factor-binding protein-1 & 2 (IGFBP-1, IGFBP-2) and (c) developmental endothelial locus 1 (DEL1) and milk fat globule–EGF factor-8 (MFG-E8). In addition, for our readers to appreciate the mounting evidence that a multitude of bone secreted factors affect the activity of other tissues, we provide a brief overview of other proteins, to include fibroblast growth factor 23 (FGF23), phosphatase orphan 1 (PHOSPHO1), osteocalcin (OCN/BGLAP), tissue non-specific alkaline phosphatase (TNAP) and acidic serine aspartic-rich MEPE-associated motif (ASARM), along with known/suggested functions of these factors in influencing energy metabolism.
Collapse
|
5
|
Faramia J, Hao Z, Mumphrey MB, Townsend RL, Miard S, Carreau AM, Nadeau M, Frisch F, Baraboi ED, Grenier-Larouche T, Noll C, Li M, Biertho L, Marceau S, Hould FS, Lebel S, Morrison CD, Münzberg H, Richard D, Carpentier AC, Tchernof A, Berthoud HR, Picard F. IGFBP-2 partly mediates the early metabolic improvements caused by bariatric surgery. Cell Rep Med 2021; 2:100248. [PMID: 33948578 PMCID: PMC8080239 DOI: 10.1016/j.xcrm.2021.100248] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 12/21/2020] [Accepted: 03/23/2021] [Indexed: 12/21/2022]
Abstract
Insulin-like growth factor-binding protein (IGFBP)-2 is a circulating biomarker of cardiometabolic health. Here, we report that circulating IGFBP-2 concentrations robustly increase after different bariatric procedures in humans, reaching higher levels after biliopancreatic diversion with duodenal switch (BPD-DS) than after Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG). This increase is closely associated with insulin sensitization. In mice and rats, BPD-DS and RYGB operations also increase circulating IGFBP-2 levels, which are not affected by SG or caloric restriction. In mice, Igfbp2 deficiency significantly impairs surgery-induced loss in adiposity and early improvement in insulin sensitivity but does not affect long-term enhancement in glucose homeostasis. This study demonstrates that the modulation of circulating IGFBP-2 may play a role in the early improvement of insulin sensitivity and loss of adiposity brought about by bariatric surgery.
Collapse
Affiliation(s)
- Justine Faramia
- Centre de recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec–Université Laval, Québec, QC, Canada
- Faculty of Pharmacy, Université Laval, Québec, QC, Canada
| | - Zheng Hao
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Michael B. Mumphrey
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - R. Leigh Townsend
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | | | - Anne-Marie Carreau
- Division of Endocrinology, Department of Medicine, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Mélanie Nadeau
- Centre de recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec–Université Laval, Québec, QC, Canada
| | - Frédérique Frisch
- Division of Endocrinology, Department of Medicine, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Elena-Dana Baraboi
- Centre de recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec–Université Laval, Québec, QC, Canada
| | - Thomas Grenier-Larouche
- Division of Endocrinology, Department of Medicine, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Christophe Noll
- Division of Endocrinology, Department of Medicine, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Meng Li
- Centre de recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec–Université Laval, Québec, QC, Canada
- Faculty of Pharmacy, Université Laval, Québec, QC, Canada
| | - Laurent Biertho
- Centre de recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec–Université Laval, Québec, QC, Canada
| | - Simon Marceau
- Centre de recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec–Université Laval, Québec, QC, Canada
| | - Frédéric-Simon Hould
- Centre de recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec–Université Laval, Québec, QC, Canada
| | - Stéfane Lebel
- Centre de recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec–Université Laval, Québec, QC, Canada
| | - Christopher D. Morrison
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Heike Münzberg
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Denis Richard
- Faculty of Pharmacy, Université Laval, Québec, QC, Canada
| | - André C. Carpentier
- Division of Endocrinology, Department of Medicine, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - André Tchernof
- Centre de recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec–Université Laval, Québec, QC, Canada
| | - Hans-Rudolf Berthoud
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Frédéric Picard
- Centre de recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec–Université Laval, Québec, QC, Canada
- Faculty of Pharmacy, Université Laval, Québec, QC, Canada
| |
Collapse
|
6
|
Cheng Z, Yin J, Yuan H, Jin C, Zhang F, Wang Z, Liu X, Wu Y, Wang T, Xiao S. Blood-Derived Plasma Protein Biomarkers for Alzheimer's Disease in Han Chinese. Front Aging Neurosci 2018; 10:414. [PMID: 30618720 PMCID: PMC6305130 DOI: 10.3389/fnagi.2018.00414] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 11/30/2018] [Indexed: 11/13/2022] Open
Abstract
It is well known that Alzheimer's disease (AD) is one of the most common progressive neurodegenerative diseases; it begins gradually, and therefore no effective medicine is administered in the beginning. Thus, early diagnosis and prevention of AD are crucial. The present study focused on comparing the plasma protein changes between patients with AD and their healthy counterparts, aiming to explore a specific protein panel as a potential biomarker for AD patients in Han Chinese. Hence, we recruited and collected plasma samples from 98 AD patients and 101 elderly healthy controls from Wuxi and Shanghai Mental Health Centers. Using a Luminex assay, we investigated the expression levels of fifty plasma proteins in these samples. Thirty-two out of 50 proteins were found to be significantly different between AD patients and healthy controls (P < 0.05). Furthermore, an eight-protein panel that included brain-derived neurotrophic factor (BDNF), angiotensinogen (AGT), insulin-like growth factor binding protein 2 (IGFBP-2), osteopontin (OPN), cathepsin D, serum amyloid P component (SAP), complement C4, and prealbumin (transthyretin, TTR) showed the highest determinative score for AD and healthy controls (all P = 0.00). In conclusion, these findings suggest that a combination of eight plasma proteins can serve as a promising diagnostic biomarker for AD with high sensitivity and specificity in Han Chinese populations; the eight plasma proteins were proven important for AD diagnosis by further cross-validation studies within the AD cohort.
Collapse
Affiliation(s)
- Zaohuo Cheng
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Jiajun Yin
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Hongwei Yuan
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Chunhui Jin
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Fuquan Zhang
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Zhiqiang Wang
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Xiaowei Liu
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Yue Wu
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Tao Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shifu Xiao
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
McLimans KE, Webb JL, Anantharam V, Kanthasamy A, Willette AA. Peripheral versus Central Index of Metabolic Dysfunction and Associations with Clinical and Pathological Outcomes in Alzheimer's Disease. J Alzheimers Dis 2018; 60:1313-1324. [PMID: 28968233 DOI: 10.3233/jad-170263] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND/OBJECTIVE Insulin-like growth factor binding protein 2 (IGFBP-2) regulates blood glucose levels, facilitates hippocampal synaptic plasticity and may have a predictive value for Alzheimer's disease (AD) diagnosis. METHODS IGFBP-2 levels were studied in plasma in 566 subjects and in cerebrospinal fluid (CSF) in 245 subjects across the AD spectrum from the Alzheimer's Disease Neuroimaging Initiative (ADNI). Variants in the IGFBP-2 gene were examined. Linear mixed modeling in SPSS tested main effects of IGFBP-2 and interactions with APOE4 on neurocognitive indices and biomarkers. Voxel-wise regression was used to gauge IGFBP-2 and regional grey matter and glucose metabolism associations. RESULTS Each point increase in IGFBP-2 corresponded to a three times greater likelihood of having mild cognitive impairment (MCI) or AD. IGFBP-2 showed beneficial associations with respect to cognitive scores in individuals with two APOE4 alleles. Higher IGFBP-2 predicted higher insulin resistance, but not CSF amyloid or tau. Voxel-wise analyses showed that plasma IGFBP-2 predicted lower grey matter volume and FDG metabolism in a large area spanning the frontal, temporal, and occipital lobes. CSF IGFBP-2 levels showed similar voxel-wise analysis results, but were uniquely associated with CSF amyloid and tau. Analysis of single nucleotide polymorphisms (SNPs) in IGFBP-2 showed that subjects carrying risk alleles versus common alleles had increased risk of AD and lower memory scores. Voxel-wise analyses of these SNPs also implicated the hippocampus and prefrontal cortex. CONCLUSIONS IGFBP-2 is associated with AD risk and outcomes; plasma IGFBP-2 provides stronger predictive power for brain outcomes, while CSF IGFBP-2 provides improved predictive accuracy for AD CSF biomarkers.
Collapse
Affiliation(s)
- Kelsey E McLimans
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA
| | - Joseph L Webb
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA
| | - Vellareddy Anantharam
- Department of Biomedical Sciences, Iowa State University, Ames, IA, USA.,Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA, USA
| | - Anumantha Kanthasamy
- Department of Biomedical Sciences, Iowa State University, Ames, IA, USA.,Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA, USA
| | - Auriel A Willette
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA.,Department of Biomedical Sciences, Iowa State University, Ames, IA, USA.,Neuroscience Graduate Program, Iowa State University, Ames, IA, USA.,Department of Psychology, Iowa State University, Ames, IA, USA.,Department of Neurology, University of Iowa, Iowa City, IA, USA
| | | |
Collapse
|
8
|
Abstract
Insulin-like growth factor-binding proteins (IGFBPs) 1-6 bind IGFs but not insulin with high affinity. They were initially identified as serum carriers and passive inhibitors of IGF actions. However, subsequent studies showed that, although IGFBPs inhibit IGF actions in many circumstances, they may also potentiate these actions. IGFBPs are widely expressed in most tissues, and they are flexible endocrine and autocrine/paracrine regulators of IGF activity, which is essential for this important physiological system. More recently, individual IGFBPs have been shown to have IGF-independent actions. Mechanisms underlying these actions include (i) interaction with non-IGF proteins in compartments including the extracellular space and matrix, the cell surface and intracellular space, (ii) interaction with and modulation of other growth factor pathways including EGF, TGF-β and VEGF, and (iii) direct or indirect transcriptional effects following nuclear entry of IGFBPs. Through these IGF-dependent and IGF-independent actions, IGFBPs modulate essential cellular processes including proliferation, survival, migration, senescence, autophagy and angiogenesis. They have been implicated in a range of disorders including malignant, metabolic, neurological and immune diseases. A more complete understanding of their cellular roles may lead to the development of novel IGFBP-based therapeutic opportunities.
Collapse
Affiliation(s)
- L A Bach
- Department of Medicine (Alfred)Monash University, Melbourne, Australia
- Department of Endocrinology and DiabetesAlfred Hospital, Melbourne, Australia
| |
Collapse
|
9
|
Clemmons DR. Role of IGF-binding proteins in regulating IGF responses to changes in metabolism. J Mol Endocrinol 2018; 61:T139-T169. [PMID: 29563157 DOI: 10.1530/jme-18-0016] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 03/21/2018] [Indexed: 12/22/2022]
Abstract
The IGF-binding protein family contains six members that share significant structural homology. Their principal function is to regulate the actions of IGF1 and IGF2. These proteins are present in plasma and extracellular fluids and regulate access of both IGF1 and II to the type I IGF receptor. Additionally, they have functions that are independent of their ability to bind IGFs. Each protein is regulated independently of IGF1 and IGF2, and this provides an important mechanism by which other hormones and physiologic variables can regulate IGF actions indirectly. Several members of the family are sensitive to changes in intermediary metabolism. Specifically the presence of obesity/insulin resistance can significantly alter the expression of these proteins. Similarly changes in nutrition or catabolism can alter their synthesis and degradation. Multiple hormones such as glucocorticoids, androgens, estrogen and insulin regulate IGFBP synthesis and bioavailability. In addition to their ability to regulate IGF access to receptors these proteins can bind to distinct cell surface proteins or proteins in extracellular matrix and several cellular functions are influenced by these interactions. IGFBPs can be transported intracellularly and interact with nuclear proteins to alter cellular physiology. In pathophysiologic states, there is significant dysregulation between the changes in IGFBP synthesis and bioavailability and changes in IGF1 and IGF2. These discordant changes can lead to marked alterations in IGF action. Although binding protein physiology and pathophysiology are complex, experimental results have provided an important avenue for understanding how IGF actions are regulated in a variety of physiologic and pathophysiologic conditions.
Collapse
Affiliation(s)
- David R Clemmons
- Department of MedicineUNC School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
10
|
Al Qahtani A, Holly J, Perks C. Hypoxia negates hyperglycaemia-induced chemo-resistance in breast cancer cells: the role of insulin-like growth factor binding protein 2. Oncotarget 2017; 8:74635-74648. [PMID: 29088813 PMCID: PMC5650368 DOI: 10.18632/oncotarget.20287] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 07/25/2017] [Indexed: 12/11/2022] Open
Abstract
Background Women who suffer from breast cancer and type II diabetes with associated hyperglycaemia respond less well to chemotherapy. We have shown that hyperglycaemia induces resistance to chemotherapy through upregulation of fatty acid synthase (FASN) in breast cancer cells and increased insulin-like binding protein 2 (IGFBP-2) in prostate cancer cells. As a tumour develops the tumour mass can outgrow the blood supply resulting in the cancer cells being exposed to hypoxia that stimulates many tumorigenic signalling pathways. Methods We used MCF-7 and T47D breast cancer cell lines. Trypan blue dye exclusion assay was employed to assess cell death and Western immunoblotting was used to determine changes in protein abundance. Hypoxia was induced both chemically by the addition of cobalt chloride (CoCl2) and using a hypoxia chamber. Results IGFBP-2 abundance increased with increasing concentrations of glucose (0-25 mM) that contributed to hyperglycaemia-induced chemo-resistance as it was abrogated by downregulating IGFBP-2 using siRNA. Production of IGFBP-2 is ER dependent: pre-treatment of MCF-7 cells with β-estradiol increased IGFBP-2 and induced chemo-resistance to doxorubicin. The hyperglycaemia-induced chemo-resistance and increases in FASN and IGFBP-2 were negated in a hypoxic environment, with levels of cell death unaffected by glucose concentrations. Conclusions The sensitivity of breast cancer cells to chemotherapy is reduced in hyperglycaemic conditions but this effect is negated by hypoxia. These effects appear to be mediated via regulation of IGFBP-2 and FASN. Understanding the role of FASN and IGFBP-2 in chemo-resistance could provide a novel target for improving the effectiveness of breast cancer treatment.
Collapse
Affiliation(s)
- Athba Al Qahtani
- IGFs and Metabolic Endocrinology Group, School of Clinical Sciences, University of Bristol, Learning and Research Building, Southmead Hospital, Bristol BS10 1TD, UK
| | - Jeff Holly
- IGFs and Metabolic Endocrinology Group, School of Clinical Sciences, University of Bristol, Learning and Research Building, Southmead Hospital, Bristol BS10 1TD, UK
| | - Claire Perks
- IGFs and Metabolic Endocrinology Group, School of Clinical Sciences, University of Bristol, Learning and Research Building, Southmead Hospital, Bristol BS10 1TD, UK
| |
Collapse
|
11
|
Schindler N, Mayer J, Saenger S, Gimsa U, Walz C, Brenmoehl J, Ohde D, Wirthgen E, Tuchscherer A, Russo VC, Frank M, Kirschstein T, Metzger F, Hoeflich A. Phenotype analysis of male transgenic mice overexpressing mutant IGFBP-2 lacking the Cardin-Weintraub sequence motif: Reduced expression of synaptic markers and myelin basic protein in the brain and a lower degree of anxiety-like behaviour. Growth Horm IGF Res 2017; 33:1-8. [PMID: 27919008 DOI: 10.1016/j.ghir.2016.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 10/18/2016] [Accepted: 11/14/2016] [Indexed: 01/07/2023]
Abstract
Brain growth and function are regulated by insulin-like growth factors I and II (IGF-I and IGF-II) but also by IGF-binding proteins (IGFBPs), including IGFBP-2. In addition to modulating IGF activities, IGFBP-2 interacts with a number of components of the extracellular matrix and cell membrane via a Cardin-Weintraub sequence or heparin binding domain (HBD1). The nature and the signalling elicited by these interactions are not fully understood. Here, we examined transgenic mice (H1d-hBP2) overexpressing a mutant human IGFBP-2 that lacks a specific heparin binding domain (HBD1) known as the Cardin-Weintraub sequence. H1d-hBP2 transgenic mice have the genetic background of FVB mice and are characterized by severe deficits in brain growth throughout their lifetime (p<0.05). In tissue lysates from brain hemispheres of 12-21day old male mice, protein levels of the GTPase dynamin-I were significantly reduced (p<0.01). Weight reductions were also found in distinct brain regions in two different age groups (12 and 80weeks). In the younger group, impaired weights were observed in the hippocampus (-34%; p<0.001), cerebellum (-25%; p<0.0001), olfactory bulb (-31%; p<0.05) and prefrontal cortex (-29%; p<0.05). At an age of 12weeks expression of myelin basic protein was reduced (p<0.01) in H1d-BP-2 mice in the cerebellum but not in the hippocampus. At 80weeks of age, weight reductions were similarly present in the cerebellum (-28%; p<0.001) and hippocampus (-31; p<0.05). When mice were challenged in the elevated plus maze, aged but not younger H1d-hBP2 mice displayed significantly less anxiety-like behaviour, which was also observed in a second transgenic mouse model overexpressing mouse IGFBP-2 lacking HBD1 (H1d-mBP2). These in vivo studies provide, for the first time, evidence for a specific role of IGFBP-2 in brain functions associated with anxiety and risk behaviour. These activities of IGFBP-2 could be mediated by the Cardin-Weintraub/HBD1 sequence and are altered in mice expressing IGFBP-2 lacking the HBD1.
Collapse
Affiliation(s)
- N Schindler
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - J Mayer
- Oscar Langendorff Institute of Physiology, University of Rostock, Germany
| | - S Saenger
- F. Hoffmann-La Roche AG, pRED, Pharma Research & Early Development, DTA CNS, Basel, Switzerland
| | - U Gimsa
- Institute of Behavioural Physiology, Leibniz-Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - C Walz
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - J Brenmoehl
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - D Ohde
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - E Wirthgen
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - A Tuchscherer
- Institute of Genetic and Biometry, Leibniz-Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - V C Russo
- Hormone Research, Murdoch Childrens Research Institute, University of Melbourne, Australia
| | - M Frank
- Medical Biology and Electron Microscopy Centre, University Medicine Rostock, Rostock, Germany
| | - T Kirschstein
- Oscar Langendorff Institute of Physiology, University of Rostock, Germany
| | - F Metzger
- F. Hoffmann-La Roche AG, pRED, Pharma Research & Early Development, DTA CNS, Basel, Switzerland
| | - A Hoeflich
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), Dummerstorf, Germany.
| |
Collapse
|
12
|
Burgdorf J, Colechio EM, Ghoreishi-Haack N, Gross AL, Rex CS, Zhang XL, Stanton PK, Kroes RA, Moskal JR. IGFBP2 Produces Rapid-Acting and Long-Lasting Effects in Rat Models of Posttraumatic Stress Disorder via a Novel Mechanism Associated with Structural Plasticity. Int J Neuropsychopharmacol 2017; 20:476-484. [PMID: 28158790 PMCID: PMC5458343 DOI: 10.1093/ijnp/pyx007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/18/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Posttraumatic stress disorder is an anxiety disorder characterized by deficits in the extinction of aversive memories. Insulin-like growth factor 1 (IGF1) is the only growth factor that has shown anxiolytic and antidepressant properties in human clinical trials. In animal studies, insulin-like growth factor binding protein 2 (IGFBP2) shows both IGF1-dependent and IGF1-independent pharmacological effects, and IGFBP2 expression is upregulated by rough-and-tumble play that induces resilience to stress. METHODS IGFBP2 was evaluated in Porsolt, contextual fear conditioning, and chronic unpredictable stress models of posttraumatic stress disorder. The dependence of IGFBP2 effects on IGF1- and AMPA-receptor activation was tested using selective receptor antagonists. Dendritic spine morphology was measured in the dentate gyrus and the medial prefrontal cortex 24 hours after in vivo dosing. RESULTS IGFBP2 was 100 times more potent than IGF1 in the Porsolt test. Unlike IGF1, effects of IGFBP2 were not blocked by the IGF1-receptor antagonist JB1, or by the AMPA-receptor antagonist 2,3-Dioxo-6-nitro-1,2,3,4 tetrahydrobenzo[f]quinoxaline-7-sulfonamide (NBQX) in the Porsolt test. IGFBP2 (1 µg/kg) and IGF1 (100 µg/kg i.v.) each facilitated contextual fear extinction and consolidation. Using a chronic unpredictable stress paradigm, IGFBP2 reversed stress-induced effects in the Porsolt, novelty-induced hypophagia, sucrose preference, and ultrasonic vocalization assays. IGFBP2 also increased mature dendritic spine densities in the medial prefrontal cortex and hippocampus 24 hours postdosing. CONCLUSIONS These data suggest that IGFBP2 has therapeutic-like effects in multiple rat models of posttraumatic stress disorder via a novel IGF1 receptor-independent mechanism. These data also suggest that the long-lasting effects of IGFBP2 may be due to facilitation of structural plasticity at the dendritic spine level. IGFBP2 and mimetics may have therapeutic potential for the treatment of posttraumatic stress disorder.
Collapse
Affiliation(s)
- Jeffrey Burgdorf
- Falk Center for Molecular Therapeutics, Department of Biomedical Engineering, Northwestern University, Evanston, Illinois (Drs Burgdorf and Moskal); Aptinyx Inc., Evanston, Illinois (Dr Colechio, Ms Ghoreishi-Haack, and Drs Gross, Kroes, and Moskal); Afraxis Inc., La Jolla, California (Dr Rex); Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York (Drs Zhang and Stanton)
| | - Elizabeth M. Colechio
- Falk Center for Molecular Therapeutics, Department of Biomedical Engineering, Northwestern University, Evanston, Illinois (Drs Burgdorf and Moskal); Aptinyx Inc., Evanston, Illinois (Dr Colechio, Ms Ghoreishi-Haack, and Drs Gross, Kroes, and Moskal); Afraxis Inc., La Jolla, California (Dr Rex); Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York (Drs Zhang and Stanton)
| | - Nayereh Ghoreishi-Haack
- Falk Center for Molecular Therapeutics, Department of Biomedical Engineering, Northwestern University, Evanston, Illinois (Drs Burgdorf and Moskal); Aptinyx Inc., Evanston, Illinois (Dr Colechio, Ms Ghoreishi-Haack, and Drs Gross, Kroes, and Moskal); Afraxis Inc., La Jolla, California (Dr Rex); Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York (Drs Zhang and Stanton)
| | - Amanda L. Gross
- Falk Center for Molecular Therapeutics, Department of Biomedical Engineering, Northwestern University, Evanston, Illinois (Drs Burgdorf and Moskal); Aptinyx Inc., Evanston, Illinois (Dr Colechio, Ms Ghoreishi-Haack, and Drs Gross, Kroes, and Moskal); Afraxis Inc., La Jolla, California (Dr Rex); Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York (Drs Zhang and Stanton)
| | - Christopher S. Rex
- Falk Center for Molecular Therapeutics, Department of Biomedical Engineering, Northwestern University, Evanston, Illinois (Drs Burgdorf and Moskal); Aptinyx Inc., Evanston, Illinois (Dr Colechio, Ms Ghoreishi-Haack, and Drs Gross, Kroes, and Moskal); Afraxis Inc., La Jolla, California (Dr Rex); Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York (Drs Zhang and Stanton)
| | - Xiao-lei Zhang
- Falk Center for Molecular Therapeutics, Department of Biomedical Engineering, Northwestern University, Evanston, Illinois (Drs Burgdorf and Moskal); Aptinyx Inc., Evanston, Illinois (Dr Colechio, Ms Ghoreishi-Haack, and Drs Gross, Kroes, and Moskal); Afraxis Inc., La Jolla, California (Dr Rex); Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York (Drs Zhang and Stanton)
| | - Patric K. Stanton
- Falk Center for Molecular Therapeutics, Department of Biomedical Engineering, Northwestern University, Evanston, Illinois (Drs Burgdorf and Moskal); Aptinyx Inc., Evanston, Illinois (Dr Colechio, Ms Ghoreishi-Haack, and Drs Gross, Kroes, and Moskal); Afraxis Inc., La Jolla, California (Dr Rex); Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York (Drs Zhang and Stanton)
| | - Roger A. Kroes
- Falk Center for Molecular Therapeutics, Department of Biomedical Engineering, Northwestern University, Evanston, Illinois (Drs Burgdorf and Moskal); Aptinyx Inc., Evanston, Illinois (Dr Colechio, Ms Ghoreishi-Haack, and Drs Gross, Kroes, and Moskal); Afraxis Inc., La Jolla, California (Dr Rex); Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York (Drs Zhang and Stanton)
| | - Joseph R. Moskal
- Falk Center for Molecular Therapeutics, Department of Biomedical Engineering, Northwestern University, Evanston, Illinois (Drs Burgdorf and Moskal); Aptinyx Inc., Evanston, Illinois (Dr Colechio, Ms Ghoreishi-Haack, and Drs Gross, Kroes, and Moskal); Afraxis Inc., La Jolla, California (Dr Rex); Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York (Drs Zhang and Stanton)
| |
Collapse
|
13
|
Swiderski K, Martins KJB, Chee A, Trieu J, Naim T, Gehrig SM, Baum DM, Brenmoehl J, Chau L, Koopman R, Gregorevic P, Metzger F, Hoeflich A, Lynch GS. Skeletal muscle-specific overexpression of IGFBP-2 promotes a slower muscle phenotype in healthy but not dystrophic mdx mice and does not affect the dystrophic pathology. Growth Horm IGF Res 2016; 30-31:1-10. [PMID: 27544574 DOI: 10.1016/j.ghir.2016.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/25/2016] [Accepted: 07/27/2016] [Indexed: 01/14/2023]
Abstract
OBJECTIVE The insulin-like growth factor binding proteins (IGFBPs) are thought to modulate cell size and homeostasis via IGF-I-dependent and -independent pathways. There is a considerable dearth of information regarding the function of IGFBPs in skeletal muscle, particularly their role in the pathophysiology of Duchenne muscular dystrophy (DMD). In this study we tested the hypothesis that intramuscular IGFBP-2 overexpression would ameliorate the pathology in mdx dystrophic mice. DESIGN 4week old male C57Bl/10 and mdx mice received a single intramuscular injection of AAV6-empty or AAV6-IGFBP-2 vector into the tibialis anterior muscle. At 8weeks post-injection the effect of IGFBP-2 overexpression on the structure and function of the injected muscle was assessed. RESULTS AAV6-mediated IGFBP-2 overexpression in the tibialis anterior (TA) muscles of 4-week-old C57BL/10 and mdx mice reduced the mass of injected muscle after 8weeks, inducing a slower muscle phenotype in C57BL/10 but not mdx mice. Analysis of inflammatory and fibrotic gene expression revealed no changes between control and IGFBP-2 injected muscles in dystrophic (mdx) mice. CONCLUSIONS Together these results indicate that the IGFBP-2-induced promotion of a slower muscle phenotype is impaired in muscles of dystrophin-deficient mdx mice, which contributes to the inability of IGFBP-2 to ameliorate the dystrophic pathology. The findings implicate the dystrophin-glycoprotein complex (DGC) in the signaling required for this adaptation.
Collapse
Affiliation(s)
- Kristy Swiderski
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, VIC 3010, Australia
| | - Karen Janet Bernice Martins
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, VIC 3010, Australia
| | - Annabel Chee
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, VIC 3010, Australia
| | - Jennifer Trieu
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, VIC 3010, Australia
| | - Timur Naim
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, VIC 3010, Australia
| | - Stefan Martin Gehrig
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, VIC 3010, Australia
| | - Dale Michael Baum
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, VIC 3010, Australia
| | - Julia Brenmoehl
- Institute for Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| | - Luong Chau
- Institute for Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| | - René Koopman
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, VIC 3010, Australia
| | - Paul Gregorevic
- Muscle Biology and Therapeutics Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Friedrich Metzger
- F. Hoffmann-La Roche Ltd., pRED, Pharma Research & Early Development, DTA Neuroscience, 4070, Basel, Switzerland
| | - Andreas Hoeflich
- Institute for Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| | - Gordon Stuart Lynch
- Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, VIC 3010, Australia.
| |
Collapse
|
14
|
Clemmons DR. Role of IGF Binding Proteins in Regulating Metabolism. Trends Endocrinol Metab 2016; 27:375-391. [PMID: 27117513 DOI: 10.1016/j.tem.2016.03.019] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 03/31/2016] [Accepted: 03/31/2016] [Indexed: 01/10/2023]
Abstract
Insulin-like growth factors (IGFs) circulate in extracellular fluids bound to a family of binding proteins. Although they function in a classical manner to limit the access of the IGFs to their receptors they also have a multiplicity of actions that are independent of this property; they bind to their own receptors or are transported to intracellular and intranuclear sites to influence cellular functions that may directly or indirectly modify IGF actions. The availability of genetically modified animals has helped to determine their functions in a physiological context. These results show that many of their actions are cell type- and context-specific, and have led to a broader understanding of how these proteins function coordinately with IGF-I and -II to regulate growth and metabolism.
Collapse
Affiliation(s)
- David R Clemmons
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA.
| |
Collapse
|
15
|
Hoeflich A, Reyer A, Ohde D, Schindler N, Brenmoehl J, Spitschak M, Langhammer M, Tuchscherer A, Wirthgen E, Renner‐Müller I, Wanke R, Metzger F, Bielohuby M, Wolf E. Dissociation of somatic growth, time of sexual maturity, and life expectancy by overexpression of an RGD-deficient IGFBP-2 variant in female transgenic mice. Aging Cell 2016; 15:111-7. [PMID: 26507795 PMCID: PMC4717279 DOI: 10.1111/acel.12413] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2015] [Indexed: 12/20/2022] Open
Abstract
Impaired growth is often associated with an extension of lifespan. However, the negative correlation between somatic growth and life expectancy is only true within, but not between, species. This can be observed because smaller species have, as a rule, a shorter lifespan than larger species. In insects and worms, reduced reproductive development and increased fat storage are associated with prolonged lifespan. However, in mammals the relationship between the dynamics of reproductive development, fat metabolism, growth rate, and lifespan are less clear. To address this point, female transgenic mice that were overexpressing similar levels of either intact (D‐mice) or mutant insulin‐like growth factor‐binding protein‐2 (IGFBP‐2) lacking the Arg‐Gly‐Asp (RGD) motif (E‐ mice) were investigated. Both lines of transgenic mice exhibited a similar degree of growth impairment (−9% and −10%) in comparison with wild‐type controls (C‐mice). While in D‐mice, sexual maturation was found to be delayed and life expectancy was significantly increased in comparison with C‐mice, these parameters were unaltered in E‐mice in spite of their reduced growth rate. These observations indicate that the RGD‐domain has a major influence on the pleiotropic effects of IGFBP‐2 and suggest that somatic growth and time of sexual maturity or somatic growth and life expectancy are less closely related than thought previously.
Collapse
Affiliation(s)
- Andreas Hoeflich
- Institute for Genome Biology Leibniz Institute for Farm Animal Biology (FBN) 18196 Dummerstorf Germany
| | - Anja Reyer
- Institute for Genome Biology Leibniz Institute for Farm Animal Biology (FBN) 18196 Dummerstorf Germany
| | - Daniela Ohde
- Institute for Genome Biology Leibniz Institute for Farm Animal Biology (FBN) 18196 Dummerstorf Germany
| | - Nancy Schindler
- Institute for Genome Biology Leibniz Institute for Farm Animal Biology (FBN) 18196 Dummerstorf Germany
| | - Julia Brenmoehl
- Institute for Genome Biology Leibniz Institute for Farm Animal Biology (FBN) 18196 Dummerstorf Germany
| | - Marion Spitschak
- Institute for Genome Biology Leibniz Institute for Farm Animal Biology (FBN) 18196 Dummerstorf Germany
| | - Martina Langhammer
- Institute for Genetics and Biometry Leibniz Institute for Farm Animal Biology (FBN) 18196 Dummerstorf Germany
| | - Armin Tuchscherer
- Institute for Genetics and Biometry Leibniz Institute for Farm Animal Biology (FBN) 18196 Dummerstorf Germany
| | - Elisa Wirthgen
- Institute for Genome Biology Leibniz Institute for Farm Animal Biology (FBN) 18196 Dummerstorf Germany
- Ligandis GbR 18276 Gülzow‐Prüzen Germany
| | - Ingrid Renner‐Müller
- Institute of Molecular Animal Breeding and Biotechnology Gene Center LMU Munich 81377 Munich Germany
| | - Rüdiger Wanke
- Institute of Veterinary Pathology LMU Munich 80539 Munich Germany
| | - Friedrich Metzger
- F. Hoffmann‐La Roche Ltd. pRED Pharma Research & Early Development DTA Neuroscience 4070 Basel Switzerland
| | - Maximilian Bielohuby
- Endocrine Research Unit Medizinische Klinik und Poliklinik IV Klinikum der Universität 80336 Munich Germany
| | - Eckhard Wolf
- Institute of Molecular Animal Breeding and Biotechnology Gene Center LMU Munich 81377 Munich Germany
- German Center for Diabetes Research (DZD) 85764 Neuherberg Germany
| |
Collapse
|