1
|
Miao J, Zhang Y, Su C, Zheng Q, Guo J. Insulin-Like Growth Factor Signaling in Alzheimer's Disease: Pathophysiology and Therapeutic Strategies. Mol Neurobiol 2025; 62:3195-3225. [PMID: 39240280 PMCID: PMC11790777 DOI: 10.1007/s12035-024-04457-1] [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: 04/24/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia among the elderly population, posing a significant public health challenge due to limited therapeutic options that merely delay cognitive decline. AD is associated with impaired energy metabolism and reduced neurotrophic signaling. The insulin-like growth factor (IGF) signaling pathway, crucial for central nervous system (CNS) development, metabolism, repair, cognition, and emotion regulation, includes IGF-1, IGF-2, IGF-1R, IGF-2R, insulin receptor (IR), and six insulin-like growth factor binding proteins (IGFBPs). Research has identified abnormalities in IGF signaling in individuals with AD and AD models. Dysregulated expression of IGFs, receptors, IGFBPs, and disruptions in downstream phosphoinositide 3-kinase-protein kinase B (PI3K/AKT) and mitogen-activated protein kinase (MAPK) pathways collectively increase AD susceptibility. Studies suggest modulating the IGF pathway may ameliorate AD pathology and cognitive decline. This review explores the CNS pathophysiology of IGF signaling in AD progression and assesses the potential of targeting the IGF system as a novel therapeutic strategy. Further research is essential to elucidate how aberrant IGF signaling contributes to AD development, understand underlying molecular mechanisms, and evaluate the safety and efficacy of IGF-based treatments.
Collapse
Affiliation(s)
- Jie Miao
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yanli Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
- Department of Neurology, Sixth Hospital of Shanxi Medical University (General Hospital of Tisco), Taiyuan, 030001, Shanxi, China
| | - Chen Su
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Qiandan Zheng
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Junhong Guo
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
2
|
Alshaalan KS, Albawardi TK, Zhra M, Bin Sulaiman N, Jnied OY, Saleem RA, Aljada A. Differential Expression of LMNA/C and Insulin Receptor Transcript Variants in Peripheral Blood Mononuclear Cells of Leukemia Patients. J Clin Med 2024; 13:2568. [PMID: 38731097 PMCID: PMC11084221 DOI: 10.3390/jcm13092568] [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: 03/18/2024] [Revised: 04/16/2024] [Accepted: 04/21/2024] [Indexed: 05/13/2024] Open
Abstract
Background: Recent research has identified alternative transcript variants of LMNA/C (LMNA, LMNC, LMNAΔ10, and LMNAΔ50) and insulin receptors (INSRs) as potential biomarkers for various types of cancer. The objective of this study was to assess the expression of LMNA/C and INSR transcript variants in peripheral blood mononuclear cells (PBMCs) of leukemia patients to investigate their potential as diagnostic biomarkers. Methods: Quantitative TaqMan reverse transcriptase polymerase chain reaction (RT-qPCR) was utilized to quantify the mRNA levels of LMNA/C (LMNA, LMNC, LMNAΔ10, and LMNAΔ50) as well as INSR (IR-A and IR-B) variants in PBMCs obtained from healthy individuals (n = 32) and patients diagnosed with primary leukemias (acute myeloid leukemia (AML): n = 17; acute lymphoblastic leukemia (ALL): n = 8; chronic myeloid leukemia (CML): n = 5; and chronic lymphocytic leukemia (CLL): n = 15). Results: Only LMNA and LMNC transcripts were notably present in PBMCs. Both exhibited significantly decreased expression levels in leukemia patients compared to the healthy control group. Particularly, the LMNC:LMNA ratio was notably higher in AML patients. Interestingly, IR-B expression was not detectable in any of the PBMC samples, precluding the calculation of the IR-A:IR-B ratio as a diagnostic marker. Despite reduced expression across all types of leukemia, IR-A levels remained detectable, indicating its potential involvement in disease progression. Conclusions: This study highlights the distinct expression patterns of LMNA/C and INSR transcript variants in PBMCs of leukemia patients. The LMNC:LMNA ratio shows promise as a potential diagnostic indicator for AML, while further research is necessary to understand the role of IR-A in leukemia pathogenesis and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Khalid Saud Alshaalan
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| | - Turki Khalid Albawardi
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| | - Mahmoud Zhra
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.Z.)
| | - Norah Bin Sulaiman
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.Z.)
| | - Osama Yaheia Jnied
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.Z.)
| | - Rimah Abdullah Saleem
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.Z.)
| | - Ahmad Aljada
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.Z.)
| |
Collapse
|
3
|
Li J, Huang G. Insulin receptor alternative splicing in breast and prostate cancer. Cancer Cell Int 2024; 24:62. [PMID: 38331804 PMCID: PMC10851471 DOI: 10.1186/s12935-024-03252-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/01/2024] [Indexed: 02/10/2024] Open
Abstract
Cancer etiology represents an intricate, multifactorial orchestration where metabolically associated insulin-like growth factors (IGFs) and insulin foster cellular proliferation and growth throughout tumorigenesis. The insulin receptor (IR) exhibits two splice variants arising from alternative mRNA processing, namely IR-A, and IR-B, with remarkable distribution and biological effects disparities. This insightful review elucidates the structural intricacies, widespread distribution, and functional significance of IR-A and IR-B. Additionally, it explores the regulatory mechanisms governing alternative splicing processes, intricate signal transduction pathways, and the intricate association linking IR-A and IR-B splicing variants to breast and prostate cancer tumorigenesis. Breast cancer and prostate cancer are the most common malignant tumors with the highest incidence rates among women and men, respectively. These findings provide a promising theoretical framework for advancing preventive strategies, diagnostic modalities, and therapeutic interventions targeting breast and prostate cancer.
Collapse
Affiliation(s)
- Jinyu Li
- Department of Medical Oncology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian, 116023, Liaoning, China
| | - Gena Huang
- Department of Medical Oncology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian, 116023, Liaoning, China.
| |
Collapse
|
4
|
Galal MA, Alouch SS, Alsultan BS, Dahman H, Alyabis NA, Alammar SA, Aljada A. Insulin Receptor Isoforms and Insulin Growth Factor-like Receptors: Implications in Cell Signaling, Carcinogenesis, and Chemoresistance. Int J Mol Sci 2023; 24:15006. [PMID: 37834454 PMCID: PMC10573852 DOI: 10.3390/ijms241915006] [Citation(s) in RCA: 11] [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: 08/21/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
This comprehensive review thoroughly explores the intricate involvement of insulin receptor (IR) isoforms and insulin-like growth factor receptors (IGFRs) in the context of the insulin and insulin-like growth factor (IGF) signaling (IIS) pathway. This elaborate system encompasses ligands, receptors, and binding proteins, giving rise to a wide array of functions, including aspects such as carcinogenesis and chemoresistance. Detailed genetic analysis of IR and IGFR structures highlights their distinct isoforms, which arise from alternative splicing and exhibit diverse affinities for ligands. Notably, the overexpression of the IR-A isoform is linked to cancer stemness, tumor development, and resistance to targeted therapies. Similarly, elevated IGFR expression accelerates tumor progression and fosters chemoresistance. The review underscores the intricate interplay between IRs and IGFRs, contributing to resistance against anti-IGFR drugs. Consequently, the dual targeting of both receptors could present a more effective strategy for surmounting chemoresistance. To conclude, this review brings to light the pivotal roles played by IRs and IGFRs in cellular signaling, carcinogenesis, and therapy resistance. By precisely modulating these receptors and their complex signaling pathways, the potential emerges for developing enhanced anti-cancer interventions, ultimately leading to improved patient outcomes.
Collapse
Affiliation(s)
- Mariam Ahmed Galal
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 1QU, UK
| | - Samhar Samer Alouch
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Buthainah Saad Alsultan
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Huda Dahman
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Nouf Abdullah Alyabis
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Sarah Ammar Alammar
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Ahmad Aljada
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
5
|
Castillo-Sanchez R, Cortes-Reynosa P, Lopez-Perez M, Garcia-Hernandez A, Salazar EP. Caveolae Microdomains Mediate STAT5 Signaling Induced by Insulin in MCF-7 Breast Cancer Cells. J Membr Biol 2023; 256:79-90. [PMID: 35751654 DOI: 10.1007/s00232-022-00253-x] [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: 02/21/2022] [Accepted: 06/06/2022] [Indexed: 02/07/2023]
Abstract
Caveolae are small plasma membrane invaginations constituted for membrane proteins namely caveolins and cytosolic proteins termed cavins, which can occupy up to 50% of the surface of mammalian cells. The caveolae have been involved with a variety of cellular processes including regulation of cellular signaling. Insulin is a hormone that mediates a variety of physiological processes through activation of insulin receptor (IR), which is a tyrosine kinase receptor expressed in all mammalian tissues. Insulin induces activation of signal transducers and activators of transcription (STAT) family members including STAT5. In this study, we demonstrate, for the first time, that insulin induces phosphorylation of STAT5 at tyrosine-694 (STAT5-Tyr(P)694), STAT5 nuclear accumulation and an increase in STAT5-DNA complex formation in MCF-7 breast cancer cells. Insulin also induces nuclear accumulation of STAT5-Tyr(P)694, caveolin-1, and IR in MCF-7 cells. STAT5 nuclear accumulation and the increase of STAT5-DNA complex formation require the integrity of caveolae and microtubule network. Moreover, insulin induces an increase and nuclear accumulation of STAT5-Tyr(P)694 in MDA-MB-231 breast cancer cells. In conclusion, results demonstrate that caveolae and microtubule network play an important role in STAT5-Tyr(P)694, STAT5 nuclear accumulation and STAT5-DNA complex formation induced by insulin in breast cancer cells.
Collapse
Affiliation(s)
- Rocio Castillo-Sanchez
- Departamento de Biologia Celular, Cinvestav-IPN, Av. IPN # 2508, 07360, Mexico City, Mexico
| | - Pedro Cortes-Reynosa
- Departamento de Biologia Celular, Cinvestav-IPN, Av. IPN # 2508, 07360, Mexico City, Mexico
| | - Mario Lopez-Perez
- Departamento de Biologia Celular, Cinvestav-IPN, Av. IPN # 2508, 07360, Mexico City, Mexico
| | | | - Eduardo Perez Salazar
- Departamento de Biologia Celular, Cinvestav-IPN, Av. IPN # 2508, 07360, Mexico City, Mexico.
| |
Collapse
|
6
|
Subventricular zone adult mouse neural stem cells require insulin receptor for self-renewal. Stem Cell Reports 2022; 17:1411-1427. [PMID: 35523180 PMCID: PMC9213826 DOI: 10.1016/j.stemcr.2022.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 11/25/2022] Open
Abstract
The insulin receptor (INSR) is an evolutionarily conserved signaling protein that regulates development and cellular metabolism. INSR signaling promotes neurogenesis in Drosophila; however, a specific role for the INSR in maintaining adult neural stem cells (NSCs) in mammals has not been investigated. We show that conditionally deleting the Insr gene in adult mouse NSCs reduces subventricular zone NSCs by ∼70% accompanied by a corresponding increase in progenitors. Insr deletion also produced hyposmia caused by aberrant olfactory bulb neurogenesis. Interestingly, hippocampal neurogenesis and hippocampal-dependent behaviors were unperturbed. Highly aggressive proneural and mesenchymal glioblastomas had high INSR/insulin-like growth factor (IGF) pathway gene expression, and isolated glioma stem cells had an aberrantly high ratio of INSR:IGF type 1 receptor. Moreover, INSR knockdown inhibited GBM tumorsphere growth. Altogether, these data demonstrate that the INSR is essential for a subset of normal NSCs, as well as for brain tumor stem cell self-renewal. Insulin receptor (INSR) is essential for adult SVZ neural stem cell self-renewal INSR deletion causes hyposmia with increased olfactory bulb neurogenesis Hippocampal stem cells (and associated behaviors) do not require INSR Glioblastomas overexpress INSR pathway components required for tumorsphere growth
Collapse
|
7
|
Insulin Resistance and Cancer: In Search for a Causal Link. Int J Mol Sci 2021; 22:ijms222011137. [PMID: 34681797 PMCID: PMC8540232 DOI: 10.3390/ijms222011137] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022] Open
Abstract
Insulin resistance (IR) is a condition which refers to individuals whose cells and tissues become insensitive to the peptide hormone, insulin. Over the recent years, a wealth of data has made it clear that a synergistic relationship exists between IR, type 2 diabetes mellitus, and cancer. Although the underlying mechanism(s) for this association remain unclear, it is well established that hyperinsulinemia, a hallmark of IR, may play a role in tumorigenesis. On the other hand, IR is strongly associated with visceral adiposity dysfunction and systemic inflammation, two conditions which favor the establishment of a pro-tumorigenic environment. Similarly, epigenetic modifications, such as DNA methylation, histone modifications, and non-coding RNA, in IR states, have been often associated with tumorigenesis in numerous types of human cancer. In addition to these observations, it is also broadly accepted that gut microbiota may play an intriguing role in the development of IR-related diseases, including type 2 diabetes and cancer, whereas potential chemopreventive properties have been attributed to some of the most commonly used antidiabetic medications. Herein we provide a concise overview of the most recent literature in this field and discuss how different but interrelated molecular pathways may impact on tumor development.
Collapse
|
8
|
Huang G, Song C, Wang N, Qin T, Sui S, Obr A, Zeng L, Wood TL, Leroith D, Li M, Wu Y. RNA-binding protein CUGBP1 controls the differential INSR splicing in molecular subtypes of breast cancer cells and affects cell aggressiveness. Carcinogenesis 2020; 41:1294-1305. [PMID: 31958132 PMCID: PMC7513956 DOI: 10.1093/carcin/bgz141] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/18/2019] [Accepted: 12/11/2019] [Indexed: 12/15/2022] Open
Abstract
The insulin receptor gene (INSR) undergoes alternative splicing to give rise to two functionally related, but also distinct, isoforms IR-A and IR-B, which dictate proliferative and metabolic regulations, respectively. Previous studies identified the RNA-binding protein CUGBP1 as a key regulator of INSR splicing. In this study, we show that the differential splicing of INSR occurs more frequently in breast cancer than in non-tumor breast tissues. In breast cancer cell lines, the IR-A:IR-B ratio varies in different molecular subtypes, knockdown or overexpression of CUGBP1 gene in breast cancer cells altered IR-A:IR-B ratio through modulation of IR-A expression, thereby reversed or enhanced the insulin-induced oncogenic behavior of breast cancer cells, respectively. Our data revealed the predominant mitogenic role of IR-A isoform in breast cancer and depicted a novel interplay between INSR and CUGBP1, implicating CUGBP1 and IR-A isoform as the potential therapeutic targets and biomarkers for breast cancer.
Collapse
Affiliation(s)
- Gena Huang
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China
- Department of Breast Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
- National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provence Key Lab of Genome Engineered Animal Models, Dalian Medical University, Dalian, Liaoning, China
| | - Chen Song
- Department of Breast Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Ning Wang
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China
- National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provence Key Lab of Genome Engineered Animal Models, Dalian Medical University, Dalian, Liaoning, China
| | - Tao Qin
- Department of Pathology, Dalian Medical University, Dalian, Liaoning, China
| | - Silei Sui
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Alison Obr
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Cancer Institute of New Jersey, Newark, NJ, USA
| | - Li Zeng
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China
- National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning, China
| | - Teresa L Wood
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Cancer Institute of New Jersey, Newark, NJ, USA
| | - Derek Leroith
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn Mount Sinai School of Medicine, New York, NY, USA
| | - Man Li
- Department of Breast Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yingjie Wu
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China
- National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provence Key Lab of Genome Engineered Animal Models, Dalian Medical University, Dalian, Liaoning, China
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn Mount Sinai School of Medicine, New York, NY, USA
- College of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
9
|
Onogi Y, Khalil AEMM, Ussar S. Identification and characterization of adipose surface epitopes. Biochem J 2020; 477:2509-2541. [PMID: 32648930 PMCID: PMC7360119 DOI: 10.1042/bcj20190462] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 12/14/2022]
Abstract
Adipose tissue is a central regulator of metabolism and an important pharmacological target to treat the metabolic consequences of obesity, such as insulin resistance and dyslipidemia. Among the various cellular compartments, the adipocyte cell surface is especially appealing as a drug target as it contains various proteins that when activated or inhibited promote adipocyte health, change its endocrine function and eventually maintain or restore whole-body insulin sensitivity. In addition, cell surface proteins are readily accessible by various drug classes. However, targeting individual cell surface proteins in adipocytes has been difficult due to important functions of these proteins outside adipose tissue, raising various safety concerns. Thus, one of the biggest challenges is the lack of adipose selective surface proteins and/or targeting reagents. Here, we discuss several receptor families with an important function in adipogenesis and mature adipocytes to highlight the complexity at the cell surface and illustrate the problems with identifying adipose selective proteins. We then discuss that, while no unique adipocyte surface protein might exist, how splicing, posttranslational modifications as well as protein/protein interactions can create enormous diversity at the cell surface that vastly expands the space of potentially unique epitopes and how these selective epitopes can be identified and targeted.
Collapse
Affiliation(s)
- Yasuhiro Onogi
- RG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Ahmed Elagamy Mohamed Mahmoud Khalil
- RG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Siegfried Ussar
- RG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
- Department of Medicine, Technische Universität München, Munich, Germany
| |
Collapse
|
10
|
Mate A, Blanca AJ, Salsoso R, Toledo F, Stiefel P, Sobrevia L, Vázquez CM. Insulin Therapy in Pregnancy Hypertensive Diseases and its Effect on the Offspring and Mother Later in Life. Curr Vasc Pharmacol 2020; 17:455-464. [PMID: 30426902 DOI: 10.2174/1570161117666181114125109] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 10/07/2018] [Accepted: 10/10/2018] [Indexed: 01/24/2023]
Abstract
Pregnancy hypertensive disorders such as Preeclampsia (PE) are strongly correlated with insulin resistance, a condition in which the metabolic handling of D-glucose is deficient. In addition, the impact of preeclampsia is enhanced by other insulin-resistant disorders, including polycystic ovary syndrome and obesity. For this reason, there is a clear association between maternal insulin resistance, polycystic ovary syndrome, obesity and the development of PE. However, whether PE is a consequence or the cause of these disorders is still unclear. Insulin therapy is usually recommended to pregnant women with diabetes mellitus when dietary and lifestyle measures have failed. The advantage of insulin therapy for Gestational Diabetes Mellitus (GDM) patients with hypertension is still controversial; surprisingly, there are no studies in which insulin therapy has been used in patients with hypertension in pregnancy without or with an established GDM. This review is focused on the use of insulin therapy in hypertensive disorders in the pregnancy and its effect on offspring and mother later in life. PubMed and relevant medical databases have been screened for literature covering research in the field especially in the last 5-10 years.
Collapse
Affiliation(s)
- Alfonso Mate
- Departamento de Fisiologia, Facultad de Farmacia, Universidad de Sevilla, E-41012 Sevilla, Spain.,Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/ Universidad de Sevilla, E- 41013 Sevilla, Spain
| | - Antonio J Blanca
- Departamento de Fisiologia, Facultad de Farmacia, Universidad de Sevilla, E-41012 Sevilla, Spain
| | - Rocío Salsoso
- Departamento de Fisiologia, Facultad de Farmacia, Universidad de Sevilla, E-41012 Sevilla, Spain.,Unidad de Enfermedades Coronarias Agudas, Instituto del Corazón, Escuela de Medicina, Universidad de Sao Paulo, Sao Paulo 05403-000 Brazil
| | - Fernando Toledo
- Department of Basic Sciences, Faculty of Sciences, Universidad del Bío-Bío, Chillan 3780000, Chile.,Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontifical Catholic University of Chile, Santiago 8330024, Chile
| | - Pablo Stiefel
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/ Universidad de Sevilla, E- 41013 Sevilla, Spain
| | - Luis Sobrevia
- Departamento de Fisiologia, Facultad de Farmacia, Universidad de Sevilla, E-41012 Sevilla, Spain.,Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontifical Catholic University of Chile, Santiago 8330024, Chile.,University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, QLD 4029, Queensland, Australia
| | - Carmen M Vázquez
- Departamento de Fisiologia, Facultad de Farmacia, Universidad de Sevilla, E-41012 Sevilla, Spain.,Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/ Universidad de Sevilla, E- 41013 Sevilla, Spain
| |
Collapse
|
11
|
Osher E, Macaulay VM. Therapeutic Targeting of the IGF Axis. Cells 2019; 8:E895. [PMID: 31416218 PMCID: PMC6721736 DOI: 10.3390/cells8080895] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/04/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022] Open
Abstract
The insulin like growth factor (IGF) axis plays a fundamental role in normal growth and development, and when deregulated makes an important contribution to disease. Here, we review the functions mediated by ligand-induced IGF axis activation, and discuss the evidence for the involvement of IGF signaling in the pathogenesis of cancer, endocrine disorders including acromegaly, diabetes and thyroid eye disease, skin diseases such as acne and psoriasis, and the frailty that accompanies aging. We discuss the use of IGF axis inhibitors, focusing on the different approaches that have been taken to develop effective and tolerable ways to block this important signaling pathway. We outline the advantages and disadvantages of each approach, and discuss progress in evaluating these agents, including factors that contributed to the failure of many of these novel therapeutics in early phase cancer trials. Finally, we summarize grounds for cautious optimism for ongoing and future studies of IGF blockade in cancer and non-malignant disorders including thyroid eye disease and aging.
Collapse
Affiliation(s)
- Eliot Osher
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | | |
Collapse
|
12
|
Vella V, Malaguarnera R, Nicolosi ML, Morrione A, Belfiore A. Insulin/IGF signaling and discoidin domain receptors: An emerging functional connection. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:118522. [PMID: 31394114 DOI: 10.1016/j.bbamcr.2019.118522] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 12/28/2022]
Abstract
The insulin/insulin-like growth factor system (IIGFs) plays a fundamental role in the regulation of prenatal and postnatal growth, metabolism and homeostasis. As a consequence, dysregulation of this axis is associated with growth disturbance, type 2 diabetes, chronic inflammation and tumor progression. A functional crosstalk between IIGFs and discoidin domain receptors (DDRs) has been recently discovered. DDRs are non-integrin collagen receptors that canonically undergo slow and long-lasting autophosphorylation after binding to fibrillar collagen. While both DDR1 and DDR2 functionally interact with IIGFs, the crosstalk with DDR1 is so far better characterized. Notably, the IIGFs-DDR1 crosstalk presents a feed-forward mechanism, which does not require collagen binding, thus identifying novel non-canonical action of DDR1. Further studies are needed to fully explore the role of this IIGFs-DDRs functional loop as potential target in the treatment of inflammatory and neoplastic disorders.
Collapse
Affiliation(s)
- Veronica Vella
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, Catania, Italy
| | | | - Maria Luisa Nicolosi
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, Catania, Italy
| | - Andrea Morrione
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Antonino Belfiore
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, Catania, Italy.
| |
Collapse
|
13
|
Wu D, Lv D, Zhang T, Guo L, Ma F, Zhang C, Lv G, Huang L. Antitumor effects of β-elemene via targeting the phosphorylation of insulin receptor. Endocr Relat Cancer 2019; 26:187-199. [PMID: 30422809 PMCID: PMC6347285 DOI: 10.1530/erc-18-0370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 11/12/2018] [Indexed: 12/20/2022]
Abstract
Ewing sarcoma family tumors (ESFTs) are a group of aggressive and highly metastatic tumors lacking efficient therapies. Insulin-like growth factor 1 receptor (IGF1R) blockade is one of the most efficient targeting therapy for ESFTs. However, the appliance is obstructed by drug resistance and disease recurrence due to the activation of insulin receptor (IR) signaling induced by IGF1R blockade. Herein β-elemene, a compound derived from natural plants, exhibited a remarkable proliferation repression on ESFT cells, which was weakened by a caspase inhibitor Z-VAD. β-elemene in combination with IGF1R inhibitors enhanced markedly the repression on cellular proliferation and mTOR activation by IGF1R inhibitors and suppressed the PI3K phosphorylation induced by IGF1R inhibitors. To investigate the mechanisms, we focused on the effects of β-elemene on IR signaling pathway. β-elemene significantly suppressed the insulin-driven cell growth and the activation of mTOR and PI3K in tumor cells, while the toxicity to normal hepatocytes was much lower. Further, the phosphorylation of IR was found to be suppressed notably by β-elemene specifically in tumor cells other than normal hepatocytes. In addition, β-elemene inhibited the growth of ESFT xenografts in vivo, and the phosphorylation of IR and S6 ribosomal protein was significantly repressed in the β-elemene-treated xenografts. These data suggest that β-elemene targets IR phosphorylation to inhibit the proliferation of tumor cells specifically and enhance the effects of IGF1R inhibitors. Thus, this study provides evidence for novel approaches by β-elemene alone or in combination with IGF1R blockades in ESFTs and IR signaling hyperactivated tumors.
Collapse
Affiliation(s)
- Dawei Wu
- Department of Pathophysiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Dongwei Lv
- Department of Sports Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Ting Zhang
- Department of Pathophysiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Lianying Guo
- Department of Pathophysiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Fangli Ma
- Department of Pathophysiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Caihua Zhang
- Department of Pathophysiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Guofeng Lv
- Department of Sports Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
- Correspondence should be addressed to L Huang or G Lv: or
| | - Lin Huang
- Department of Pathophysiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
- Correspondence should be addressed to L Huang or G Lv: or
| |
Collapse
|
14
|
Oncofoetal insulin receptor isoform A marks the tumour endothelium; an underestimated pathway during tumour angiogenesis and angiostatic treatment. Br J Cancer 2018; 120:218-228. [PMID: 30559346 PMCID: PMC6342959 DOI: 10.1038/s41416-018-0347-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/17/2018] [Accepted: 10/24/2018] [Indexed: 12/29/2022] Open
Abstract
Background In a genomic screen for determinants of the tumour vasculature, we identified insulin receptor (INSR) to mark the tumour endothelium. As a functional role for insulin/INSR in cancer has been suggested and markers of the tumour endothelium may be attractive therapeutic targets, we investigated the role of INSR in angiogenesis. Methods In a genomic screen for determinants of the tumour vasculature we identified insulin receptor to mark the tumour endothelium. Results The current report demonstrates the following: (i) the heavy overexpression of INSR on angiogenic vasculature in human tumours and the correlation to short survival, (ii) that INSR expression in the tumour vasculature is mainly representing the short oncofoetal and non-metabolic isoform INSR-A, (iii) the angiogenic activity of insulin on endothelial cells (EC) in vitro and in vivo, (iv) suppression of proliferation and sprouting of EC in vitro after antibody targeting or siRNA knockdown, and (v) inhibition of in vivo angiogenesis in the chicken chorioallantoic membrane (CAM) by anti-INSR antibodies. We additionally show, using preclinical mouse as well as patient data, that treatment with the inhibitor sunitinib significantly reduces the expression of INSR-A. Conclusions The current study underscores the oncogenic impact of INSR and suggests that targeting the INSR-A isoform should be considered in therapeutic settings.
Collapse
|
15
|
Vella V, Milluzzo A, Scalisi NM, Vigneri P, Sciacca L. Insulin Receptor Isoforms in Cancer. Int J Mol Sci 2018; 19:ijms19113615. [PMID: 30453495 PMCID: PMC6274710 DOI: 10.3390/ijms19113615] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/05/2018] [Accepted: 11/13/2018] [Indexed: 12/17/2022] Open
Abstract
The insulin receptor (IR) mediates both metabolic and mitogenic effects especially when overexpressed or in clinical conditions with compensatory hyperinsulinemia, due to the metabolic pathway resistance, as obesity diabetes. In many cancers, IR is overexpressed preferentially as IR-A isoform, derived by alternative splicing of exon 11. The IR-A overexpression, and the increased IR-A:IR-B ratio, are mechanisms that promote the mitogenic response of cancer cells to insulin and IGF-2, which is produced locally by both epithelial and stromal cancer cells. In cancer IR-A, isoform predominance may occur for dysregulation at both mRNA transcription and post-transcription levels, including splicing factors, non-coding RNAs and protein degradation. The mechanisms that regulate IR isoform expression are complex and not fully understood. The IR isoform overexpression may play a role in cancer cell stemness, in tumor progression and in resistance to target therapies. From a clinical point of view, the IR-A overexpression in cancer may be a determinant factor for the resistance to IGF-1R target therapies for this issue. IR isoform expression in cancers may have the meaning of a predictive biomarker and co-targeting IGF-1R and IR-A may represent a new more efficacious treatment strategy.
Collapse
Affiliation(s)
- Veronica Vella
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, Garibaldi-Nesima Hospital, via Palermo 636, 95122 Catania, Italy.
- School of Human and Social Science, University "Kore" of Enna, 94100 Enna, Italy.
| | - Agostino Milluzzo
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, Garibaldi-Nesima Hospital, via Palermo 636, 95122 Catania, Italy.
| | - Nunzio Massimo Scalisi
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, Garibaldi-Nesima Hospital, via Palermo 636, 95122 Catania, Italy.
| | - Paolo Vigneri
- Department of Clinical and Experimental Medicine, University of Catania Medical School, Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, via Santa Sofia, 78, 95123 Catania, Italy.
| | - Laura Sciacca
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, Garibaldi-Nesima Hospital, via Palermo 636, 95122 Catania, Italy.
| |
Collapse
|
16
|
Underrated enemy - from nonalcoholic fatty liver disease to cancers of the gastrointestinal tract. Clin Exp Hepatol 2018; 4:55-71. [PMID: 29904722 PMCID: PMC6000748 DOI: 10.5114/ceh.2018.75955] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 04/17/2018] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is intrahepatic ectopic lipid deposition which is present despite a lack of other causes of secondary hepatic fat accumulation. It is the most common chronic liver disorder in the welldeveloped countries. NAFLD is a multidisciplinary disease that affects various systems and organs and is inextricably linked to simple obesity, metabolic syndrome, insulin resistance and overt diabetes mellitus type 2. The positive energy balance related to obesity leads to a variety of systemic changes including modified levels of insulin, insulin- like growth factor-1, adipokines, hepatokines and cytokines. It is strongly linked to carcinogenesis and new evidence proves that NAFLD is associated with higher risk of all-cause mortality and cancer-specific mortality among cancer survivors. This article focuses on the association between NAFLD and extrahepatic gastrointestinal tract cancers, aiming to shed light on the pathomechanism of changes leading to the development of tumors.
Collapse
|
17
|
Belfiore A, Malaguarnera R, Vella V, Lawrence MC, Sciacca L, Frasca F, Morrione A, Vigneri R. Insulin Receptor Isoforms in Physiology and Disease: An Updated View. Endocr Rev 2017; 38:379-431. [PMID: 28973479 PMCID: PMC5629070 DOI: 10.1210/er.2017-00073] [Citation(s) in RCA: 266] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/13/2017] [Indexed: 02/08/2023]
Abstract
The insulin receptor (IR) gene undergoes differential splicing that generates two IR isoforms, IR-A and IR-B. The physiological roles of IR isoforms are incompletely understood and appear to be determined by their different binding affinities for insulin-like growth factors (IGFs), particularly for IGF-2. Predominant roles of IR-A in prenatal growth and development and of IR-B in metabolic regulation are well established. However, emerging evidence indicates that the differential expression of IR isoforms may also help explain the diversification of insulin and IGF signaling and actions in various organs and tissues by involving not only different ligand-binding affinities but also different membrane partitioning and trafficking and possibly different abilities to interact with a variety of molecular partners. Of note, dysregulation of the IR-A/IR-B ratio is associated with insulin resistance, aging, and increased proliferative activity of normal and neoplastic tissues and appears to sustain detrimental effects. This review discusses novel information that has generated remarkable progress in our understanding of the physiology of IR isoforms and their role in disease. We also focus on novel IR ligands and modulators that should now be considered as an important strategy for better and safer treatment of diabetes and cancer and possibly other IR-related diseases.
Collapse
Affiliation(s)
- Antonino Belfiore
- Endocrinology, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Roberta Malaguarnera
- Endocrinology, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Veronica Vella
- School of Human and Social Sciences, University Kore of Enna, via della Cooperazione, 94100 Enna, Italy
| | - Michael C. Lawrence
- Structural Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Laura Sciacca
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy
| | - Francesco Frasca
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy
| | - Andrea Morrione
- Department of Urology and Biology of Prostate Cancer Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Riccardo Vigneri
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy
| |
Collapse
|
18
|
Escribano O, Beneit N, Rubio-Longás C, López-Pastor AR, Gómez-Hernández A. The Role of Insulin Receptor Isoforms in Diabetes and Its Metabolic and Vascular Complications. J Diabetes Res 2017; 2017:1403206. [PMID: 29201918 PMCID: PMC5671728 DOI: 10.1155/2017/1403206] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 09/12/2017] [Accepted: 09/25/2017] [Indexed: 12/18/2022] Open
Abstract
The insulin receptor (IR) presents by alternative splicing two isoforms: IRA and IRB. The differential physiological and pathological role of both isoforms is not completely known, and it is determinant the different binding affinity for insulin-like growth factor. IRB is more abundant in adult tissues and it exerts mainly the metabolic actions of insulin, whereas IRA is mainly expressed in fetal and prenatal period and exerts mitogenic actions. However, the change in the expression profile of both IR isoforms and its dysregulation are associated with the development of different pathologies, such as cancer, insulin resistance, diabetes, obesity, and atherosclerosis. In some of them, there is a significant increase of IRA/IRB ratio conferring a proliferative and migratory advantage to different cell types and favouring IGF-II actions with a sustained detriment in the metabolic effects of insulin. This review discussed specifically the role of IR isoforms as well as IGF-IR in diabetes and its associated complications as obesity and atherosclerosis. Future research with new IR modulators might be considered as possible targets to improve the treatment of diabetes and its associated complications.
Collapse
Affiliation(s)
- O. Escribano
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Madrid, Spain
- Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain
- CIBER of Diabetes and Associated Metabolic Diseases, Madrid, Spain
| | - N. Beneit
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Madrid, Spain
- Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain
- CIBER of Diabetes and Associated Metabolic Diseases, Madrid, Spain
| | - C. Rubio-Longás
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Madrid, Spain
| | - A. R. López-Pastor
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Madrid, Spain
| | - A. Gómez-Hernández
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Madrid, Spain
- Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain
- CIBER of Diabetes and Associated Metabolic Diseases, Madrid, Spain
| |
Collapse
|
19
|
Beneit N, Fernández-García CE, Martín-Ventura JL, Perdomo L, Escribano Ó, Michel JB, García-Gómez G, Fernández S, Díaz-Castroverde S, Egido J, Gómez-Hernández A, Benito M. Expression of insulin receptor (IR) A and B isoforms, IGF-IR, and IR/IGF-IR hybrid receptors in vascular smooth muscle cells and their role in cell migration in atherosclerosis. Cardiovasc Diabetol 2016; 15:161. [PMID: 27905925 PMCID: PMC5134076 DOI: 10.1186/s12933-016-0477-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 11/22/2016] [Indexed: 01/02/2023] Open
Abstract
Background Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) is a major contributor to the development of atherosclerotic process. In a previous work, we demonstrated that the insulin receptor isoform A (IRA) and its association with the insulin-like growth factor-I receptor (IGF-IR) confer a proliferative advantage to VSMCs. However, the role of IR and IGF-IR in VSMC migration remains poorly understood. Methods Wound healing assays were performed in VSMCs bearing IR (IRLoxP+/+ VSMCs), or not (IR−/− VSMCs), expressing IRA (IRA VSMCs) or expressing IRB (IRB VSMCs). To study the role of IR isoforms and IGF-IR in experimental atherosclerosis, we used ApoE−/− mice at 8, 12, 18 and 24 weeks of age. Finally, we analyzed the mRNA expression of total IR, IRB isoform, IGF-IR and IGFs by qRT-PCR in the medial layer of human aortas. Results IGF-I strongly induced migration of the four cell lines through IGF-IR. In contrast, insulin and IGF-II only caused a significant increase of IRA VSMC migration which might be favored by the formation of IRA/IGF-IR receptors. Additionally, a specific IGF-IR inhibitor, picropodophyllin, completely abolished insulin- and IGF-II-induced migration in IRB, but not in IRA VSMCs. A significant increase of IRA and IGF-IR, and VSMC migration were observed in fibrous plaques from 24-week-old ApoE−/− mice. Finally, we observed a marked increase of IGF-IR, IGF-I and IGF-II in media from fatty streaks as compared with both healthy aortas and fibrolipidic lesions, favoring the ability of medial VSMCs to migrate into the intima. Conclusions Our data suggest that overexpression of IGF-IR or IRA isoform, as homodimers or as part of IRA/IGF-IR hybrid receptors, confers a stronger migratory capability to VSMCs as might occur in early stages of atherosclerotic process. Electronic supplementary material The online version of this article (doi:10.1186/s12933-016-0477-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- N Beneit
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - C E Fernández-García
- Vascular Research Lab, IIS-Fundación Jiménez Diaz-Autonoma University, Madrid, Spain
| | - J L Martín-Ventura
- Vascular Research Lab, IIS-Fundación Jiménez Diaz-Autonoma University, Madrid, Spain
| | - L Perdomo
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - Ó Escribano
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - J B Michel
- Inserm, U698, Universite Paris 7, CHU X-Bichat, Paris, France
| | - G García-Gómez
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - S Fernández
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - S Díaz-Castroverde
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - J Egido
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain.,Vascular Research Lab, IIS-Fundación Jiménez Diaz-Autonoma University, Madrid, Spain
| | - A Gómez-Hernández
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain. .,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain. .,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain.
| | - M Benito
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| |
Collapse
|
20
|
Westermeier F, Sáez T, Arroyo P, Toledo F, Gutiérrez J, Sanhueza C, Pardo F, Leiva A, Sobrevia L. Insulin receptor isoforms: an integrated view focused on gestational diabetes mellitus. Diabetes Metab Res Rev 2016; 32:350-65. [PMID: 26431063 DOI: 10.1002/dmrr.2729] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 09/14/2015] [Accepted: 09/27/2015] [Indexed: 12/17/2022]
Abstract
The human insulin receptor (IR) exists in two isoforms that differ by the absence (IR-A) or the presence (IR-B) of a 12-amino acid segment encoded by exon 11. Both isoforms are functionally distinct regarding their binding affinities and intracellular signalling. However, the underlying mechanisms related to their cellular functions in several tissues are only partially understood. In this review, we summarize the current knowledge in this field regarding the alternative splicing of IR isoform, tissue-specific distribution and signalling both in physiology and disease, with an emphasis on the human placenta in gestational diabetes mellitus (GDM). Furthermore, we discuss the clinical relevance of IR isoforms highlighted by findings that show altered insulin signalling due to differential IR-A and IR-B expression in human placental endothelium in GDM pregnancies. Future research and clinical studies focused on the role of IR isoform signalling might provide novel therapeutic targets for treating GDM to improve the adverse maternal and neonatal outcomes.
Collapse
Affiliation(s)
- F Westermeier
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Advanced Centre for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Faculty of Science, Universidad San Sebastián, Santiago, Chile
| | - T Sáez
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- University Medical Centre Groningen (UMCG), Faculty of Medicine, University of Groningen, Groningen, The Netherlands
| | - P Arroyo
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - F Toledo
- Department of Basic Sciences, Faculty of Sciences, Universidad del Bío-Bío, Chillán, Chile
| | - J Gutiérrez
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Cellular Signalling and Differentiation Laboratory (CSDL), School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Santiago, Chile
| | - C Sanhueza
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - F Pardo
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - A Leiva
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - L Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville, Spain
- University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Queensland, Australia
| |
Collapse
|
21
|
Ouyang H, Yang HS, Yu T, Shan TD, Li JY, Huang CZ, Zhong W, Xia ZS, Chen QK. MEK/ERK pathway activation by insulin receptor isoform alteration is associated with the abnormal proliferation and differentiation of intestinal epithelial cells in diabetic mice. Mol Cell Biochem 2016; 413:165-78. [PMID: 26724951 DOI: 10.1007/s11010-015-2650-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 12/23/2015] [Indexed: 12/15/2022]
Abstract
In previous studies, we have reported the abnormal proliferation and differentiation of intestinal epithelial cells (IECs) in diabetes mellitus (DM) mice. The insulin receptor (IR) and its downstream mitogen-activated protein kinase kinase (MAPKK also known as MEK)/extracellular-regulated protein kinase (ERK) pathway is a classic pathway associated with cell proliferation and differentiation. The purpose of the present study is to investigate the role of the MEK/ERK pathway in abnormal proliferation and differentiation of IECs in DM mice. DM mouse models were induced by intraperitoneal injection of streptozotocin. The expression levels of the IR and its isoforms in IECs of DM mice and in IEC-6 cells were investigated. To ensure that the downstream pathways were monitored, QPCR and Western blotting were performed to detect the expression levels of MEK1/2, ERK1/2, PI3K, and Akt. Moreover, siRNA for IR-A and U0126, a specific inhibitor of MEK, were used to further investigate the relationship between the IR/MEK/ERK pathway and abnormal proliferation and differentiation of IECs in DM mice. In DM mice, excessive proliferation, disturbed differentiation, and a high ratio of IR-A/IR-B were detected in IECs. The expression levels of MEK1, MEK2, and ERK1/2 and their phosphorylated proteins in DM mice were significantly higher than those in the control group (P < 0.05), which could be offset by using siRNA for IR-A. The abnormal proliferation and differentiation of IECs in DM mice were normalized after the in vivo administration of U0126. The abnormal proliferation and differentiation of IECs in DM mice are associated with high IR-A/IR-B ratio and increased IR/MEK/ERK pathway activity.
Collapse
Affiliation(s)
- Hui Ouyang
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan Jiang Xi Road, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Hong-Sheng Yang
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan Jiang Xi Road, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Tao Yu
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan Jiang Xi Road, Guangzhou, 510120, Guangdong, People's Republic of China.
| | - Ti-Dong Shan
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan Jiang Xi Road, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Jie-Yao Li
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan Jiang Xi Road, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Can-Ze Huang
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan Jiang Xi Road, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Wa Zhong
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan Jiang Xi Road, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Zhong-Sheng Xia
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan Jiang Xi Road, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Qi-Kui Chen
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan Jiang Xi Road, Guangzhou, 510120, Guangdong, People's Republic of China.
| |
Collapse
|
22
|
Obesity and cancer, a case for insulin signaling. Cell Death Dis 2015; 6:e2037. [PMID: 26720346 PMCID: PMC4720912 DOI: 10.1038/cddis.2015.381] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 11/24/2015] [Accepted: 11/26/2015] [Indexed: 02/08/2023]
Abstract
Obesity is a worldwide epidemic, with the number of overweight and obese individuals climbing from just over 500 million in 2008 to 1.9 billion in 2014. Type 2 diabetes (T2D), cardiovascular disease and non-alcoholic fatty liver disease have long been associated with the obese state, whereas cancer is quickly emerging as another pathological consequence of this disease. Globally, at least 2.8 million people die each year from being overweight or obese. It is estimated that by 2020 being overweight or obese will surpass the health burden of tobacco consumption. Increase in the body mass index (BMI) in overweight (BMI>25 kg/m2) and obese (BMI>30 kg/m2) individuals is a result of adipose tissue (AT) expansion, which can lead to fat comprising >50% of the body weight in the morbidly obese. Extensive research over the last several years has painted a very complex picture of AT biology. One clear link between AT expansion and etiology of diseases like T2D and cancer is the development of insulin resistance (IR) and hyperinsulinemia. This review focuses on defining the link between obesity, IR and cancer.
Collapse
|
23
|
Takarada T, Nishida A, Takeuchi A, Lee T, Takeshima Y, Matsuo M. Resveratrol enhances splicing of insulin receptor exon 11 in myotonic dystrophy type 1 fibroblasts. Brain Dev 2015; 37:661-8. [PMID: 25476247 DOI: 10.1016/j.braindev.2014.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 11/07/2014] [Accepted: 11/07/2014] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Myotonic dystrophy type 1 (DM1) is characterized by splicing abnormalities caused by CUG expansion of the DMPK gene transcript. Splicing of exon 11 of the insulin receptor (IR) gene is deregulated to suppress exon 11 inclusion into mRNA in DM1. Consequently, the exon 11-deleted IR isoform that is less sensitive to insulin is predominantly produced, leading to glucose intolerance in DM1. Upregulation of exon 11 retaining full-length IR mRNA is a potential way to recover insulin sensitivity in DM1. METHODS We examined candidate chemicals for their ability to enhance inclusion of exon 11 of the IR gene in cultured cells by reverse transcription-PCR amplification of a fragment extending from exons 10 to 12 of IR mRNA. RESULTS We revealed that resveratrol (RES) enhanced the percentage of exon 11-containing IR mRNA among the total IR mRNA in HeLa cells. The RES-mediated enhancement of exon 11 inclusion was cell-specific and highest in fibroblasts. We tested RES on four fibroblast samples from three generations of one DM1 family. In each sample, RES treatment significantly upregulated the percentage of exon 11-containing IR mRNA to levels higher than that of the control, irrespective of the length of the sample's CTG repeat expansion. DISCUSSION A natural compound, RES, was shown for the first time to upregulate the full-length IR mRNA in fibroblasts from DM1 cases. Our results provide the justification of RES as a leading compound to improve glucose tolerance in DM1.
Collapse
Affiliation(s)
- Toru Takarada
- Department of Medical Rehabilitation, Faculty of Rehabilitation, Kobegakuin University, Nishi, Kobe 6512180, Japan; Kobe Pharmaceutical University, Higashinada, Kobe 6588558, Japan
| | - Atsushi Nishida
- Department of Medical Rehabilitation, Faculty of Rehabilitation, Kobegakuin University, Nishi, Kobe 6512180, Japan
| | - Atsuko Takeuchi
- Kobe Pharmaceutical University, Higashinada, Kobe 6588558, Japan
| | - Tomoko Lee
- Department of Pediatrics, Hyogo College of Medicine, Nishinomiya 6638131, Japan
| | - Yasuhiro Takeshima
- Department of Pediatrics, Hyogo College of Medicine, Nishinomiya 6638131, Japan
| | - Masafumi Matsuo
- Department of Medical Rehabilitation, Faculty of Rehabilitation, Kobegakuin University, Nishi, Kobe 6512180, Japan.
| |
Collapse
|
24
|
Abstract
Neural stem cells (NSCs) are found in two regions in the adult brain: the subgranular zone (SGZ) in the hippocampal dentate gyrus and the subventricular zone (SVZ) adjacent to the lateral ventricles. Similarly to other somatic stem cells, adult NSCs are found within specialized niches that are organized to facilitate NSC self-renewal. Alterations in stem-cell homeostasis can contribute to the consequences of neurodegenerative diseases, healthy ageing and tissue repair after damage. Insulin and the insulin-like growth factors (IGFs) function in stem-cell homeostasis across species. Studies in the mammalian central nervous system support essential roles for IGF and/or insulin signalling in NSC self-renewal, neurogenesis, cognition and sensory function through distinct ligand-receptor interactions. IGF-II is of particular interest as a result of its production by the choroid plexus and presence in cerebrospinal fluid (CSF). CSF regulates and supports the development, division and migration of cells in the adult brain and is required for NSC maintenance. In this Review, we discuss emerging data on the functions of IGF-II and IGF and/or insulin receptor signalling in the context of NSC regulation in the SVZ and SGZ. We also propose a model for IGF-II in which the choroid plexus is a major component of the NSC niche.
Collapse
Affiliation(s)
- Amber N Ziegler
- Department of Neurology &Neuroscience, New Jersey Medical School, Rutgers Biomedical &Health Sciences, Cancer Centre, 205 South Orange Avenue, Newark, NJ 07101, USA
| | - Steven W Levison
- Department of Neurology &Neuroscience, New Jersey Medical School, Rutgers Biomedical &Health Sciences, Cancer Centre, 205 South Orange Avenue, Newark, NJ 07101, USA
| | - Teresa L Wood
- Department of Neurology &Neuroscience, New Jersey Medical School, Rutgers Biomedical &Health Sciences, Cancer Centre, 205 South Orange Avenue, Newark, NJ 07101, USA
| |
Collapse
|
25
|
The Individual and Combined Effects of Obesity and Type 2 Diabetes on Cancer Predisposition and Survival. Curr Nutr Rep 2015. [DOI: 10.1007/s13668-014-0105-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
26
|
De Marco P, Cirillo F, Vivacqua A, Malaguarnera R, Belfiore A, Maggiolini M. Novel Aspects Concerning the Functional Cross-Talk between the Insulin/IGF-I System and Estrogen Signaling in Cancer Cells. Front Endocrinol (Lausanne) 2015; 6:30. [PMID: 25798130 PMCID: PMC4351617 DOI: 10.3389/fendo.2015.00030] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 02/19/2015] [Indexed: 12/13/2022] Open
Abstract
The insulin/IGF system plays an important role in cancer progression. Accordingly, elevated levels of circulating insulin have been associated with an increased cancer risk as well as with aggressive and metastatic cancer phenotypes. Numerous studies have documented that estrogens cooperate with the insulin/IGF system in multiple pathophysiological conditions. The biological responses to estrogens are mainly mediated by the estrogen receptors (ER)α and ERβ, which act as transcription factors; however, several studies have recently demonstrated that a member of the G protein-coupled receptors, named GPR30/G-protein estrogen receptor (GPER), is also involved in the estrogen signaling in normal and malignant cells as well as in cancer-associated fibroblasts (CAFs). In this regard, novel mechanisms linking the action of estrogens through GPER with the insulin/IGF system have been recently demonstrated. This review recapitulates the relevant aspects of this functional cross-talk between the insulin/IGF and the estrogenic GPER transduction pathways, which occurs in various cell types and may account for cancer progression.
Collapse
Affiliation(s)
- Paola De Marco
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Francesca Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Adele Vivacqua
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Roberta Malaguarnera
- Endocrinology, Department of Health, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Antonino Belfiore
- Endocrinology, Department of Health, University Magna Graecia of Catanzaro, Catanzaro, Italy
- *Correspondence: Antonino Belfiore, Università degli Studi Magna Graecia di Catanzaro, Viale Europa, Loc. Germaneto, Catanzaro 88100, Italy e-mail: ; Marcello Maggiolini, Università della Calabria, via P. Bucci, Rende 87036, Italy e-mail:
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
- *Correspondence: Antonino Belfiore, Università degli Studi Magna Graecia di Catanzaro, Viale Europa, Loc. Germaneto, Catanzaro 88100, Italy e-mail: ; Marcello Maggiolini, Università della Calabria, via P. Bucci, Rende 87036, Italy e-mail:
| |
Collapse
|
27
|
Enguita-Germán M, Fortes P. Targeting the insulin-like growth factor pathway in hepatocellular carcinoma. World J Hepatol 2014; 6:716-737. [PMID: 25349643 PMCID: PMC4209417 DOI: 10.4254/wjh.v6.i10.716] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 07/14/2014] [Accepted: 08/31/2014] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related deaths worldwide. Only 30%-40% of the patients with HCC are eligible for curative treatments, which include surgical resection as the first option, liver transplantation and percutaneous ablation. Unfortunately, there is a high frequency of tumor recurrence after surgical resection and most HCC seem resistant to conventional chemotherapy and radiotherapy. Sorafenib, a multi-tyrosine kinase inhibitor, is the only chemotherapeutic option for patients with advanced hepatocellular carcinoma. Patients treated with Sorafenib have a significant increase in overall survival of about three months. Therefore, there is an urgent need to develop alternative treatments. Due to its role in cell growth and development, the insulin-like growth factor system is commonly deregulated in many cancers. Indeed, the insulin-like growth factor (IGF) axis has recently emerged as a potential target for hepatocellular carcinoma treatment. To this aim, several inhibitors of the pathway have been developed such as monoclonal antibodies, small molecules, antisense oligonucleotides or small interfering RNAs. However recent studies suggest that, unlike most tumors, HCC development requires increased signaling through insulin growth factor II rather than insulin growth factor I. This may have great implications in the future treatment of HCC. This review summarizes the role of the IGF axis in liver carcinogenesis and the current status of the strategies designed to target the IGF-I signaling pathway for hepatocellular carcinoma treatment.
Collapse
|
28
|
Handelsman Y, Leroith D, Bloomgarden ZT, Dagogo-Jack S, Einhorn D, Garber AJ, Grunberger G, Harrell RM, Gagel RF, Lebovitz HE, McGill JB, Hennekens CH. Diabetes and cancer--an AACE/ACE consensus statement. Endocr Pract 2014; 19:675-93. [PMID: 23978590 DOI: 10.4158/ep13248.cs] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
29
|
Malaguarnera R, Belfiore A. The emerging role of insulin and insulin-like growth factor signaling in cancer stem cells. Front Endocrinol (Lausanne) 2014; 5:10. [PMID: 24550888 PMCID: PMC3912738 DOI: 10.3389/fendo.2014.00010] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 01/21/2014] [Indexed: 12/20/2022] Open
Abstract
Cancer cells frequently exploit the IGF signaling, a fundamental pathway mediating development, cell growth, and survival. As a consequence, several components of the IGF signaling are deregulated in cancer and sustain cancer progression. However, specific targeting of IGF-IR in humans has resulted efficacious only in small subsets of cancers, making researches wondering whether IGF system targeting is still worth pursuing in the clinical setting. Although no definite answer is yet available, it has become increasingly clear that other components of the IGF signaling pathway, such as IR-A, may substitute for the lack of IGF-IR, and induce cancer resistance and/or clonal selection. Moreover, accumulating evidence now indicates that IGF signaling is a central player in the induction/maintenance of epithelial mesenchymal transition (EMT) and cell stemness, two strictly related programs, which play a key role in metastatic spread and resistance to cancer treatments. Here we review the evidences indicating that IGF signaling enhances the expression of transcription factors implicated in the EMT program and has extensive cross-talk with specific pathways involved in cell pluripotency and stemness maintenance. In turn, EMT and cell stemness activate positive feed-back mechanisms causing up-regulation of various IGF signaling components. These findings may have novel translational implications.
Collapse
Affiliation(s)
- Roberta Malaguarnera
- Endocrinology, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Antonino Belfiore
- Endocrinology, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| |
Collapse
|
30
|
Guzmán-Gutiérrez E, Arroyo P, Salsoso R, Fuenzalida B, Sáez T, Leiva A, Pardo F, Sobrevia L. Role of Insulin and Adenosine in the Human Placenta Microvascular and Macrovascular Endothelial Cell Dysfunction in Gestational Diabetes Mellitus. Microcirculation 2014; 21:26-37. [DOI: 10.1111/micc.12077] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 07/18/2013] [Indexed: 12/19/2022]
Affiliation(s)
- Enrique Guzmán-Gutiérrez
- Cellular and Molecular Physiology Laboratory (CMPL); Division of Obstetrics and Gynaecology; Medical Research Centre (CIM); School of Medicine; Faculty of Medicine; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Pablo Arroyo
- Cellular and Molecular Physiology Laboratory (CMPL); Division of Obstetrics and Gynaecology; Medical Research Centre (CIM); School of Medicine; Faculty of Medicine; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Rocío Salsoso
- Cellular and Molecular Physiology Laboratory (CMPL); Division of Obstetrics and Gynaecology; Medical Research Centre (CIM); School of Medicine; Faculty of Medicine; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Bárbara Fuenzalida
- Cellular and Molecular Physiology Laboratory (CMPL); Division of Obstetrics and Gynaecology; Medical Research Centre (CIM); School of Medicine; Faculty of Medicine; Pontificia Universidad Católica de Chile; Santiago Chile
- Biomedical Department; Faculty of Health Sciences; Universidad de Antofagasta; Antofagasta Chile
| | - Tamara Sáez
- Cellular and Molecular Physiology Laboratory (CMPL); Division of Obstetrics and Gynaecology; Medical Research Centre (CIM); School of Medicine; Faculty of Medicine; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Andrea Leiva
- Cellular and Molecular Physiology Laboratory (CMPL); Division of Obstetrics and Gynaecology; Medical Research Centre (CIM); School of Medicine; Faculty of Medicine; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Fabián Pardo
- Cellular and Molecular Physiology Laboratory (CMPL); Division of Obstetrics and Gynaecology; Medical Research Centre (CIM); School of Medicine; Faculty of Medicine; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL); Division of Obstetrics and Gynaecology; Medical Research Centre (CIM); School of Medicine; Faculty of Medicine; Pontificia Universidad Católica de Chile; Santiago Chile
- University of Queensland Centre for Clinical Research; Herston Queensland Australia
| |
Collapse
|
31
|
Wang T, Ning G, Bloomgarden Z. Diabetes and cancer relationships. J Diabetes 2013; 5:378-90. [PMID: 23574745 DOI: 10.1111/1753-0407.12057] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 03/10/2013] [Accepted: 03/24/2013] [Indexed: 12/29/2022] Open
Abstract
Diabetes and cancer are both heterogeneous and multifactorial diseases with tremendous impact on health worldwide. Epidemiologic evidence suggests that certain malignancies may be associated with diabetes, as well as with diabetes risk factors and, perhaps, with certain diabetes treatments. Numerous biological mechanisms could account for these relationships. Insulin-like growth factor (IGF)-1, IGF-2, IGF-1 receptors, insulin, and the insulin receptor play roles in the development and progression of cancers. Although evidence from randomized controlled trials does not support or refute associations of diabetes and its treatments with either increased or reduced risk of cancer incidence or prognosis, consideration of malignancy incidence rates and the magnitude of the trials that would be required to address these issues explains why such studies may not be readily undertaken.
Collapse
Affiliation(s)
- Tiange Wang
- Key Laboratory for Endocrine and Metabolic Diseases of Ministry of Health, Shanghai, China; Shanghai Clinical Center for Endocrine and Metabolic Diseases, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; E-Institute of Shanghai Universities, Shanghai, China
| | | | | |
Collapse
|
32
|
Drab SR, Philis‐Tsimikas A. A New Option for Glycemic Control: Insulin Degludec, a New‐Generation Basal Insulin with an Ultralong Duration of Action. Pharmacotherapy 2013; 34:291-302. [DOI: 10.1002/phar.1361] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Scott R. Drab
- University of Pittsburgh School of Pharmacy Pittsburgh Pennsylvania
- University Diabetes Care Associates Pittsburgh Pennsylvania
| | | |
Collapse
|
33
|
Gallagher EJ, Alikhani N, Tobin-Hess A, Blank J, Buffin NJ, Zelenko Z, Tennagels N, Werner U, LeRoith D. Insulin receptor phosphorylation by endogenous insulin or the insulin analog AspB10 promotes mammary tumor growth independent of the IGF-I receptor. Diabetes 2013; 62:3553-60. [PMID: 23835331 PMCID: PMC3781483 DOI: 10.2337/db13-0249] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Endogenous hyperinsulinemia and insulin receptor (IR)/IGF-I receptor (IGF-IR) phosphorylation in tumors are associated with a worse prognosis in women with breast cancer. In vitro, insulin stimulation of the IR increases proliferation of breast cancer cells. However, in vivo studies demonstrating that IR activation increases tumor growth, independently of IGF-IR activation, are lacking. We hypothesized that endogenous hyperinsulinemia increases mammary tumor growth by directly activating the IR rather than the IGF-IR or hybrid receptors. We aimed to determine whether stimulating the IR with the insulin analog AspB10 could increase tumor growth independently of IGF-IR signaling. We induced orthotopic mammary tumors in control FVB/n and hyperinsulinemic MKR mice, and treated them with the insulin analog AspB10, recombinant human IGF-I, or vehicle. Tumors from mice with endogenous hyperinsulinemia were larger and had greater IR phosphorylation, but not IGF-IR phosphorylation, than those from control mice. Chronic AspB10 administration also increased tumor growth and IR (but not IGF-IR) phosphorylation in tumors. IGF-I led to activation of both the IGF-IR and IR and probably hybrid receptors. Our results demonstrate that IR phosphorylation increases tumor growth, independently of IGF-IR/hybrid receptor phosphorylation, and warrant consideration when developing therapeutics targeting the IGF-IR, but not the IR.
Collapse
Affiliation(s)
- Emily Jane Gallagher
- Division of Endocrinology, Diabetes and Bone Diseases, Samuel Bronfman Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nyosha Alikhani
- Division of Endocrinology, Diabetes and Bone Diseases, Samuel Bronfman Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Aviva Tobin-Hess
- Division of Endocrinology, Diabetes and Bone Diseases, Samuel Bronfman Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jeffrey Blank
- Division of Endocrinology, Diabetes and Bone Diseases, Samuel Bronfman Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nicholas J. Buffin
- Division of Endocrinology, Diabetes and Bone Diseases, Samuel Bronfman Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Zara Zelenko
- Division of Endocrinology, Diabetes and Bone Diseases, Samuel Bronfman Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Norbert Tennagels
- R&D Diabetes Division, Sanofi-Aventis Deutschland, Frankfurt am Main, Germany
| | - Ulrich Werner
- R&D Diabetes Division, Sanofi-Aventis Deutschland, Frankfurt am Main, Germany
| | - Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Diseases, Samuel Bronfman Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Corresponding author: Derek LeRoith,
| |
Collapse
|
34
|
Gómez-Hernández A, Escribano Ó, Perdomo L, Otero YF, García-Gómez G, Fernández S, Beneit N, Benito M. Implication of insulin receptor A isoform and IRA/IGF-IR hybrid receptors in the aortic vascular smooth muscle cell proliferation: role of TNF-α and IGF-II. Endocrinology 2013; 154:2352-64. [PMID: 23677929 DOI: 10.1210/en.2012-2161] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
To assess the role of insulin receptor (IR) isoforms (IRA and IRB) in the proliferation of vascular smooth muscle cells (VSMCs) involved in the atherosclerotic process, we generated new VSMC lines bearing IR (wild-type VSMCs; IRLoxP(+/+) VSMCs), lacking IR (IR(-/-) VSMCs) or expressing IRA (IRA VSMCs) or IRB (IRB VSMCs). Insulin and different proatherogenic stimuli induced a significant increase of IRA expression in IRLoxP(+/+) VSMCs. Moreover, insulin, through ERK signaling, and the proatherogenic stimuli, through ERK and p38 signaling, induced a higher proliferation in IRA than IRB VSMCs. The latter effect might be due to IRA cells showing a higher expression of angiotensin II, endothelin 1, and thromboxane 2 receptors and basal association between IRA and these receptors. Furthermore, TNF-α induced in a ligand-dependent manner a higher association between IRA and TNF-α receptor 1 (TNF-R1). On the other hand, IRA overexpression might favor the atherogenic actions of IGF-II. Thereby, IGF-II or TNF-α induced IRA and IGF-I receptor (IGF-IR) overexpression as well as an increase of IRA/IGF-IR hybrid receptors in VSMCs. More importantly, we observed a significant increase of IRA, TNF-R1, and IGF-IR expression as well as higher association of IRA with TNF-R1 or IGF-IR in the aorta from ApoE(-/-) and BATIRKO mice, 2 models showing vascular damage. In addition, anti-TNF-α treatment prevented those effects in BATIRKO mice. Finally, our data suggest that the IRA isoform and its association with TNF-R1 or IGF-IR confers proliferative advantage to VSMCs, mainly in response to TNF-α or IGF-II, which might be of significance in the early atherosclerotic process.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Cell Proliferation/drug effects
- Cells, Cultured
- Immunoprecipitation
- Insulin-Like Growth Factor II/pharmacology
- Male
- Mice
- Mice, Knockout
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Protein Isoforms/genetics
- Protein Isoforms/metabolism
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/metabolism
- Receptor, Insulin/genetics
- Receptor, Insulin/metabolism
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- Almudena Gómez-Hernández
- Biochemistry and Molecular Biology Department, School of Pharmacy, Complutense University of Madrid, Instituto de Investigaciones Sanitarias Hospital Clínico San Carlos, Centro de Investigación Biomédica en Red of Diabetes and Associated Metabolic Diseases, Instituto de Salud Carlos III, Madrid 28040, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Jin M, Buck E, Mulvihill MJ. Modulation of insulin-like growth factor-1 receptor and its signaling network for the treatment of cancer: current status and future perspectives. Oncol Rev 2013; 7:e3. [PMID: 25992224 PMCID: PMC4419619 DOI: 10.4081/oncol.2013.e3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 04/05/2013] [Accepted: 04/15/2013] [Indexed: 12/20/2022] Open
Abstract
Based on over three decades of pre-clinical data, insulin-like growth factor-1 receptor (IGF-1R) signaling has gained recognition as a promoter of tumorogenesis, driving cell survival and proliferation in multiple human cancers. As a result, IGF-1R has been pursued as a target for cancer treatment. Early pioneering efforts targeting IGF-1R focused on highly selective monoclonal antibodies, with multiple agents advancing to clinical trials. However, despite some initial promising results, recent clinical disclosures have been less encouraging. Moreover, recent studies have revealed that IGF-1R participates in a dynamic and complex signaling network, interacting with additional targets and pathways thereof through various crosstalk and compensatory signaling mechanisms. Such mechanisms of bypass signaling help to shed some light on the decreased effectiveness of selective IGF-1R targeted therapies (e.g. monoclonal antibodies) and suggest that targeting multiple nodes within this signaling network might be necessary to produce a more effective therapeutic response. Additionally, such findings have led to the development of small molecule IGF-1R inhibitors which also co-inhibit additional targets such as insulin receptor and epidermal growth factor receptor. Such findings have helped to guide the design rationale of numerous drug combinations that are currently being evaluated in clinical trials.
Collapse
|
36
|
Palouzier-Paulignan B, Lacroix MC, Aimé P, Baly C, Caillol M, Congar P, Julliard AK, Tucker K, Fadool DA. Olfaction under metabolic influences. Chem Senses 2012; 37:769-97. [PMID: 22832483 PMCID: PMC3529618 DOI: 10.1093/chemse/bjs059] [Citation(s) in RCA: 230] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Recently published work and emerging research efforts have suggested that the olfactory system is intimately linked with the endocrine systems that regulate or modify energy balance. Although much attention has been focused on the parallels between taste transduction and neuroendocrine controls of digestion due to the novel discovery of taste receptors and molecular components shared by the tongue and gut, the equivalent body of knowledge that has accumulated for the olfactory system, has largely been overlooked. During regular cycles of food intake or disorders of endocrine function, olfaction is modulated in response to changing levels of various molecules, such as ghrelin, orexins, neuropeptide Y, insulin, leptin, and cholecystokinin. In view of the worldwide health concern regarding the rising incidence of diabetes, obesity, and related metabolic disorders, we present a comprehensive review that addresses the current knowledge of hormonal modulation of olfactory perception and how disruption of hormonal signaling in the olfactory system can affect energy homeostasis.
Collapse
Affiliation(s)
- Brigitte Palouzier-Paulignan
- Centre de Recherche des Neurosciences de Lyon, Equipe Olfaction du Codage à la Mémoire, INSERM U 1028/CNRS 5292, Université de Lyon150 Ave. Tony Garnier, 69366, Lyon, Cedex 07,France
- Equal contribution
| | - Marie-Christine Lacroix
- INRA, UR1197 Neurobiologie de l’Olfaction et Modélisation en ImagerieF-78350, Jouy-en-JosasFrance
- IFR 144NeuroSud Paris, 91190 Gif-Sur-YvetteFrance
- Equal contribution
| | - Pascaline Aimé
- Centre de Recherche des Neurosciences de Lyon, Equipe Olfaction du Codage à la Mémoire, INSERM U 1028/CNRS 5292, Université de Lyon150 Ave. Tony Garnier, 69366, Lyon, Cedex 07,France
| | - Christine Baly
- INRA, UR1197 Neurobiologie de l’Olfaction et Modélisation en ImagerieF-78350, Jouy-en-JosasFrance
- IFR 144NeuroSud Paris, 91190 Gif-Sur-YvetteFrance
| | - Monique Caillol
- INRA, UR1197 Neurobiologie de l’Olfaction et Modélisation en ImagerieF-78350, Jouy-en-JosasFrance
- IFR 144NeuroSud Paris, 91190 Gif-Sur-YvetteFrance
| | - Patrice Congar
- INRA, UR1197 Neurobiologie de l’Olfaction et Modélisation en ImagerieF-78350, Jouy-en-JosasFrance
- IFR 144NeuroSud Paris, 91190 Gif-Sur-YvetteFrance
| | - A. Karyn Julliard
- Centre de Recherche des Neurosciences de Lyon, Equipe Olfaction du Codage à la Mémoire, INSERM U 1028/CNRS 5292, Université de Lyon150 Ave. Tony Garnier, 69366, Lyon, Cedex 07,France
| | - Kristal Tucker
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of MedicinePittsburgh, PA 15261USAand
| | - Debra Ann Fadool
- Department of Biological Science, Programs in Neuroscience and Molecular Biophysics, The Florida State UniversityTallahassee, FL 32306-4295USA
| |
Collapse
|
37
|
Vigneri R, Squatrito S, Frittitta L. Selective insulin receptor modulators (SIRM): a new class of antidiabetes drugs? Diabetes 2012; 61:984-5. [PMID: 22517652 PMCID: PMC3331756 DOI: 10.2337/db12-0192] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
38
|
Avnet S, Perut F, Salerno M, Sciacca L, Baldini N. Insulin receptor isoforms are differently expressed during human osteoblastogenesis. Differentiation 2012; 83:242-8. [PMID: 22466604 DOI: 10.1016/j.diff.2012.02.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 01/27/2012] [Accepted: 02/06/2012] [Indexed: 12/13/2022]
Abstract
The reciprocal influence and bidirectional cross-talk between bone and energy metabolism is a recent finding, since the discovery that the product of osteoblasts osteocalcin increases pancreatic β-cell proliferation, insulin secretion and sensitivity. Conversely, the anabolic effect of insulin is crucial for osteoblast function, as suggested by severe osteopenia and increased incidence of fracture in insulin-deficient diabetic patients. The Insulin Receptor (IR) tyrosine kinase, which is commonly expressed in the insulin-sensitive liver, muscle, and adipose tissues, is also found in animal and human bone. Here we show that in human bone two insulin receptor isoforms (IR-A and IR-B) are differently expressed. Mature human osteoblasts predominantly express IR-B, whereas IR-A is mainly expressed in osteoblast precursors, and IR-B/IR-A mRNA ratio significantly increases along the osteogenic differentiation of mesenchymal stromal precursors. Moreover, transfected osteoprogenitors overexpressing IR-A show an increased proliferation rate. In contrast, when transfected with and overexpressing IR-B, their proliferation rate is reduced, corresponding to a more differentiated phenotype. In conclusion, the fine regulation of the expression of different isoforms of IR during osteogenic differentiation confirms the important role played by IR in bone homeostasis, providing the basis for new perspectives on the various involvements of IR isoforms in bone pathophysiology.
Collapse
Affiliation(s)
- Sofia Avnet
- Laboratory for Orthopaedic Pathophysiology and Regenerative Medicine, Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | | | | | | |
Collapse
|
39
|
Kalla Singh S, Brito C, Tan QW, De León M, De León D. Differential expression and signaling activation of insulin receptor isoforms A and B: A link between breast cancer and diabetes. Growth Factors 2011; 29:278-89. [PMID: 21913804 PMCID: PMC3205219 DOI: 10.3109/08977194.2011.616200] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We showed that when insulin-like growth factor II (IGF-II) is highly expressed in breast tissues and cell lines, the IGF-I receptor signaling pathway is highly activated. Since IGF-II activates the insulin receptor (INSR), we propose that the INSR signaling is also activated in this system. We examined the expression of both INSR isoforms, insulin receptor A (INSR-A) and insulin receptor B (INSR-B), and the downstream signaling pathways in breast cancer (BC) cells and in paired (normal/tumor) breast tissues from 100 patients. Analysis was performed by real-time PCR, Western blot, immunohistochemistry, and phospho-ELISA techniques. Tumor tissues and cell lines from African-American patients expressed higher levels of INSR-A, but lower levels of INSR-B. Accordingly, insulin receptor substrate 1 and focal adhesion kinase activation were significantly increased in these women. We conclude that higher INSR-A and lower INSR-B contribute to higher proliferation and lower metabolic response. Thus, differential expression of INSR isoforms represents a potential biological link between BC and diabetes.
Collapse
Affiliation(s)
- S Kalla Singh
- Breast Cancer Laboratory, Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, CA 92350, USA
| | | | | | | | | |
Collapse
|
40
|
Alvino CL, Ong SC, McNeil KA, Delaine C, Booker GW, Wallace JC, Forbes BE. Understanding the mechanism of insulin and insulin-like growth factor (IGF) receptor activation by IGF-II. PLoS One 2011; 6:e27488. [PMID: 22140443 PMCID: PMC3227035 DOI: 10.1371/journal.pone.0027488] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 10/18/2011] [Indexed: 12/29/2022] Open
Abstract
Background Insulin-like growth factor-II (IGF-II) promotes cell proliferation and survival and plays an important role in normal fetal development and placental function. IGF-II binds both the insulin-like growth factor receptor (IGF-1R) and insulin receptor isoform A (IR-A) with high affinity. Interestingly both IGF-II and the IR-A are often upregulated in cancer and IGF-II acts via both receptors to promote cancer proliferation. There is relatively little known about the mechanism of ligand induced activation of the insulin (IR) and IGF-1R. The recently solved IR structure reveals a folded over dimer with two potential ligand binding pockets arising from residues on each receptor half. Site-directed mutagenesis has mapped receptor residues important for ligand binding to two separate sites within the ligand binding pocket and we have recently shown that the IGFs have two separate binding surfaces which interact with the receptor sites 1 and 2. Methodology/Principal Findings In this study we describe a series of partial IGF-1R and IR agonists generated by mutating Glu12 of IGF-II. By comparing receptor binding affinities, abilities to induce negative cooperativity and potencies in receptor activation, we provide evidence that residue Glu12 bridges the two receptor halves leading to receptor activation. Conclusions/Significance This study provides novel insight into the mechanism of receptor binding and activation by IGF-II, which may be important for the future development of inhibitors of its action for the treatment of cancer.
Collapse
Affiliation(s)
- Clair L. Alvino
- School of Molecular and Biomedical Science, The University of Adelaide, Adelaide, Australia
| | - Shee Chee Ong
- School of Molecular and Biomedical Science, The University of Adelaide, Adelaide, Australia
| | - Kerrie A. McNeil
- School of Molecular and Biomedical Science, The University of Adelaide, Adelaide, Australia
| | - Carlie Delaine
- School of Molecular and Biomedical Science, The University of Adelaide, Adelaide, Australia
| | - Grant W. Booker
- School of Molecular and Biomedical Science, The University of Adelaide, Adelaide, Australia
| | - John C. Wallace
- School of Molecular and Biomedical Science, The University of Adelaide, Adelaide, Australia
| | - Briony E. Forbes
- School of Molecular and Biomedical Science, The University of Adelaide, Adelaide, Australia
- * E-mail:
| |
Collapse
|
41
|
Abstract
The widespread epidemic of obesity and type 2 diabetes has raised concern for the impact of these disorders as risk factors for cancer and has renewed the interest for studies regarding the involvement of hyperinsulinemia and insulin receptor (IR) in cancer progression. Overexpression of IR in cancer cells may explain their increased sensitivity to hyperinsulinemia. Moreover, IR isoform A (IR-A) together with autocrine production of its ligand IGF2 is emerging as an important mechanism of normal and cancer stem cell expansion and is a feature of several malignancies. De novo activation of the IR-A/IGF2 autocrine loop also represents a mechanism of resistance to anticancer therapies. Increasing knowledge of the IR role in cancer has important implications for cancer prevention, which should include control of insulin resistance and hyperinsulinemia in the population and meticulous evaluation of new antidiabetic drugs for their metabolic:mitogenic ratio. We are now aware that several anticancer treatments may induce or worsen insulin resistance that may limit therapy efficacy. Future anticancer therapies need to target the IR-A pathway in order to inhibit the tumor promoting effect of IR without impairing the metabolic effect of insulin.
Collapse
Affiliation(s)
- Antonino Belfiore
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University Magna Graecia of Catanzaro, Campus Universitario, località Germaneto, 88100 Catanzaro, Italy.
| | | |
Collapse
|
42
|
Kang HM, Park S, Kim H. Insulin-like growth factor 2 enhances insulinogenic differentiation of human eyelid adipose stem cells via the insulin receptor. Cell Prolif 2011; 44:254-63. [PMID: 21535266 DOI: 10.1111/j.1365-2184.2011.00755.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVES Previously, we have isolated stem cells (HEAC) from human eyelid adipose tissue and functionally differentiated them into insulin-secreting cells. In the present study, we examined whether insulin family members might influence insulinogenic differentiation of HEAC. MATERIALS AND METHODS Following culture in differentiation media containing insulin family member or not, cells were examined for gene expression, protein expression and, particularly, insulin and C-peptide secretion, in response to high glucose challenge. Using antibodies against the specific receptor, target receptor mediating effect of the insulin family member was investigated. RESULTS Insulin treatment during culture had little effect on either insulin or C-peptide secretion from HEAC, against high glucose challenge after culture. However, insulin-like growth factor (IGF) 1 treatment decreased both secretions, and interestingly, IGF2 greatly increased the secretions. HEAC treated with IGF2 had strong expression of Pdx1, Isl1, Pax6 and PC1/3 genes, and distinct staining after insulin and C-peptide antibodies, and dithizone. IGF2-enhanced insulinogenic differentiation was totally blocked by antibody against insulin receptor (IR), but not by anti-IGF1 receptor (IGF1R). Differentiated HEAC expressed both IR and IGF1R genes, whereas they expressed neither IGF2 nor IGF2R genes. CONCLUSIONS From these results, it is suggested that IGF1 might inhibit insulinogenic differentiation of HEAC, whereas IGF2 enhances differentiation, and that enhancement of IGF2 appeared to be mediated via IR.
Collapse
Affiliation(s)
- H M Kang
- Department of Biotechnology, Seoul Women's University, Kongnung-dong, Nowon-gu, Seoul, Korea
| | | | | |
Collapse
|
43
|
Cannata D, Fierz Y, Vijayakumar A, LeRoith D. Type 2 diabetes and cancer: what is the connection? ACTA ACUST UNITED AC 2011; 77:197-213. [PMID: 20309918 DOI: 10.1002/msj.20167] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Epidemiological studies have demonstrated an association between type 2 diabetes and cancer. Type 2 diabetes is characterized by insulin resistance and hyperinsulinemia. Hyperinsulinemia may lead to cancer through insulin's effect on its cognate receptor and the insulin-like growth factor system. The effects of insulin and insulin-like growth factor I on cancer development and progression have been demonstrated in animal and human studies. Type 2 diabetes has been positively associated with cancers of the breast, colon, and pancreas. An inverse relationship has been observed between type 2 diabetes and prostate cancer, and this may be due to lower testosterone levels in men with type 2 diabetes. Medications used to treat type 2 diabetes may affect cancer cells directly or indirectly by affecting serum insulin levels. Hyperinsulinemia may be an important risk factor for cancer as well as a target for cancer therapy.
Collapse
Affiliation(s)
- Dara Cannata
- Mount Sinai School of Medicine, New York, NY, USA
| | | | | | | |
Collapse
|
44
|
Buck E, Mulvihill M. Small molecule inhibitors of the IGF-1R/IR axis for the treatment of cancer. Expert Opin Investig Drugs 2011; 20:605-21. [PMID: 21446886 DOI: 10.1517/13543784.2011.558501] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION The IGF-1 receptor (IGF-1R) is a receptor tyrosine kinase and is well established as a key regulator of tumor cell growth and survival. There is also a growing body of data to support a role for the structurally and functionally related insulin receptor (IR) in human cancer. Bidirectional crosstalk between IGF-1R and IR is observed, where specific inhibition of either receptor confers a compensatory increase in the activity for the reciprocal receptor, therefore dual inhibition of both IGF-1R and IR may be important for optimal efficacy. The importance of IGF-1R and IR as targets in cancer is further underscored by their contribution to resistance against both cytotoxic and molecularly targeted anti-cancer therapeutics. Currently, both IGF-1R-neutralizing antibodies and small-molecule tyrosine kinase inhibitors of IGF-1R/IR are in clinical development. AREAS COVERED The importance of IGF-1R and IR as cancer targets and how IGF-1R/IR inhibitors may sensitize tumor cells to the anti-proliferative and pro-apoptotic effects of other anti-tumor agents. The potential advantages of small molecule IGF-1R/IR inhibitors compared with IGF-1R-specific neutralizing antibodies, and the characteristics of small-molecule IGF-1R inhibitors that have entered clinical development. EXPERT OPINION Because of compensatory crosstalk between IGF-1R and IR, dual IGF-1R and IR tyrosine kinase inhibitors may have superior anti-tumor activity compared to anti-IGF-1R specific antibodies. The clinical success for IGF-1R/IR inhibitors may ultimately be dependent upon our ability to correctly administer these agents to the right niche patient subpopulation using single agent therapy, when appropriate, or using the right combination therapy.
Collapse
Affiliation(s)
- Elizabeth Buck
- Translational Research, OSI Pharmaceuticals, Farmingdale, NY 11735, USA.
| | | |
Collapse
|
45
|
Malaguarnera R, Frasca F, Garozzo A, Gianì F, Pandini G, Vella V, Vigneri R, Belfiore A. Insulin receptor isoforms and insulin-like growth factor receptor in human follicular cell precursors from papillary thyroid cancer and normal thyroid. J Clin Endocrinol Metab 2011; 96:766-74. [PMID: 21123448 DOI: 10.1210/jc.2010-1255] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Factors involved in the biology of normal and cancer stem/precursor cells from the thyroid are unknown. Thyroid cancer cells are responsive to insulin and IGF-I and IGF-II and often overexpress the insulin receptor (IR) and the IGF-I receptor (IGF-IR). OBJECTIVE We investigated the role of IR isoforms (IR-A and IR-B), IGF-IR, and their ligands in thyroid follicular cell precursors both normal and malignant. DESIGN We established cultures of follicular cell precursors as thyrospheres from three papillary thyroid cancers and the corresponding nonaffected tissues. The expression of IR, IGF-IR, and their ligands was evaluated by quantitative RT-PCR and, in one case, also by Western blot. The effects of insulin and IGFs on thyrosphere growth and self-renewal were evaluated. RESULTS Thyrospheres were characterized by the expression of stem cell markers and low/absent thyroid specific markers. Thyrospheres from normal tissue, but not from cancer tissue, could be induced to differentiate. Both IR isoforms, IGF-IR, IGF-I and IGF-II, were expressed at high levels in thyrospheres and markedly decreased in differentiating cells. IR-A was the predominant isoform in thyrospheres, especially from cancer, while IR-B was predominant in differentiating cells. Cancer thyrosphere growth was stimulated by insulin and IGFs. CONCLUSIONS Our data suggest that IR isoforms and IGF-IR play a role in the biology of follicular thyroid precursors. Cell differentiation is associated with marked changes in the expression of these receptors and cognate ligands. These data may provide insight for future differentiation therapies in thyroid cancer.
Collapse
MESH Headings
- Blotting, Western
- Carcinoma, Papillary/genetics
- Carcinoma, Papillary/metabolism
- Cell Adhesion
- Cell Line, Tumor
- Cells, Cultured
- DNA Primers
- Gene Expression Regulation, Neoplastic/physiology
- Humans
- Ligands
- Neoplastic Stem Cells/metabolism
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/metabolism
- Receptor, IGF Type 2/genetics
- Receptor, IGF Type 2/metabolism
- Receptor, Insulin/genetics
- Receptor, Insulin/metabolism
- Receptors, Somatomedin/genetics
- Receptors, Somatomedin/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Thyroid Gland/cytology
- Thyroid Gland/metabolism
- Thyroid Neoplasms/genetics
- Thyroid Neoplasms/metabolism
Collapse
Affiliation(s)
- Roberta Malaguarnera
- Department of Clinical and Experimental Medicine, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Goetsch L, Corvaïa N. Insulin-like growth factor receptor type I as a target for cancer therapy. Immunotherapy 2010; 1:265-79. [PMID: 20635945 DOI: 10.2217/1750743x.1.2.265] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In recent years, improvements in the understanding of oncogenesis has permitted the identification of new molecular targets for cancer therapy. Among all the different approaches, inhibition of tyrosine kinase receptor activity using small molecules or biomolecules for controlling cancer growth has been successful and has brought new therapeutic opportunities to the medical community. After more than 20 years of extensive work, insulin-like growth factor receptor I (IGF-IR) is becoming an attractive target for drug development. Owing to its close homology to insulin receptor, IGF-IR is of interest for antibody design while its specificity allows us to discriminate between the two receptors. Major efforts from a large number of pharmaceutical companies are invested in evaluating the efficacy of such molecules in humans. Discovery of biomarkers associated with efficacy and patient selection are the main challenges that we will have to deal with in order to target the appropriate patient population that will most benefit from anti-IGF-IR monoclonal antibodies and combined treatments. This review will provide an overview of the current knowledge on IGF-IR and ongoing clinical trials.
Collapse
Affiliation(s)
- Liliane Goetsch
- Centre d'Immunologie Pierre Fabre, 5 avenue Napoléon III, F-74164 Saint Julien-en-Genevois, France.
| | | |
Collapse
|
47
|
Rosenzweig SA, Atreya HS. Defining the pathway to insulin-like growth factor system targeting in cancer. Biochem Pharmacol 2010; 80:1115-24. [PMID: 20599789 DOI: 10.1016/j.bcp.2010.06.013] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Revised: 06/12/2010] [Accepted: 06/15/2010] [Indexed: 12/28/2022]
Abstract
The insulin-like growth factors (IGFs; IGF-1 and IGF-2) play central roles in cell growth, differentiation, survival, transformation and metastasis. The biologic effects of the IGFs are mediated by the IGF-1 receptor (IGF-1R), a receptor tyrosine kinase with homology to the insulin receptor (IR). Dysregulation of the IGF system is well recognized as a key contributor to the progression of multiple cancers, with IGF-1R activation increasing the tumorigenic potential of breast, prostate, lung, colon and head and neck squamous cell carcinoma (HNSCC). Despite this relationship, targeting the IGF-1R has only recently undergone development as a molecular cancer therapeutic. As it has taken hold, we are witnessing a robust increase and interest in targeting the inhibition of IGF-1R signaling. This is accentuated by the list of over 30 drugs, including monoclonal antibodies (mAbs) and tyrosine kinase inhibitors (TKIs) that are under evaluation as single agents or in combination therapies. The IGF-binding proteins (IGFBPs) represent the third component of the IGF system consisting of a class of six soluble secretory proteins. They represent a unique class of naturally occurring IGF-antagonists that bind to and sequester IGF-1 and IGF-2, inhibiting their access to the IGF-1R. Due to their dual targeting of the IGFs without affecting insulin action, the IGFBPs are an untapped "third" class of IGF-1R inhibitors. In this commentary, we highlight some of the significant aspects of and prospects for targeting the IGF-1R and describe what the future may hold.
Collapse
Affiliation(s)
- Steven A Rosenzweig
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics and Hollings Cancer Center, Medical University of South Carolina, 173 Ashley Avenue, PO Box 250505, Charleston, SC 29425-5050, USA.
| | | |
Collapse
|
48
|
Speakman JR, Goran MI. Tissue-specificity and ethnic diversity in obesity-related risk of cancer may be explained by variability in insulin response and insulin signaling pathways. Obesity (Silver Spring) 2010; 18:1071-8. [PMID: 20150900 DOI: 10.1038/oby.2010.16] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Obesity is a predisposing risk factor for several chronic diseases. The link between obesity and cancer appears to be particularly complex. Notably only the risk for development of specific cancers appear to be affected. Moreover, the obesity-related risk of cancer is very different across ethnic groups. African-Americans appear particularly prone, whereas Hispanics appear to be relatively protected. Obesity is associated with increased levels of circulating insulin. These levels of elevated insulin may serve to promote proliferation of fat cells to accommodate the elevated nutrient flux. However, elevated levels of insulin may be a major mediating factor influencing cancer risk. This hypothesis alone cannot explain the complexity of the phenomenon. We suggest here that the different insulin responses to obesity of different ethnic groups may explain their different risk profiles. Moreover, we speculate that tissue-specific variations in the insulin signaling pathways may underlie their differential susceptibility to tumorigenesis in the face of elevated obesity. Elevated cancer risk may be an unwanted side effect of insulin responding to elevated nutrient flux in the obese which it serves to proliferate fat cells that provide a location for storage of ingested fat, which consequently prevents ectopic fat storage. Hence, while Hispanics may be protected from cancer risk in obesity because of their lower insulin response, they have an elevated risk of fatty liver disease. Reduction of insulin levels in obesity as a strategy to reduce cancer risk may pose additional problems unless it is combined also with interventions that aim to limit nutrient influx.
Collapse
Affiliation(s)
- John R Speakman
- Aberdeen Centre for Energy Regulation and Obesity (ACERO), Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, UK.
| | | |
Collapse
|
49
|
Abstract
Type 2 diabetes mellitus (T2DM) affects a large population worldwide. T2DM is a complex heterogeneous group of metabolic disorders including hyperglycemia and impaired insulin action and/or insulin secretion. T2DM causes dysfunctions in multiple organs or tissues. Current theories of T2DM include a defect in insulin-mediated glucose uptake in muscle, a dysfunction of the pancreatic beta-cells, a disruption of secretory function of adipocytes, and an impaired insulin action in liver. The etiology of human T2DM is multifactorial, with genetic background and physical inactivity as two critical components. The pathogenesis of T2DM is not fully understood. Animal models of T2DM have been proved to be useful to study the pathogenesis of, and to find a new therapy for, the disease. Although different animal models share similar characteristics, each mimics a specific aspect of genetic, endocrine, metabolic, and morphologic changes that occur in human T2DM. The purpose of this review is to provide the recent progress and current theories in T2DM and to summarize animal models for studying the pathogenesis of the disease.
Collapse
Affiliation(s)
- Yi Lin
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | | |
Collapse
|
50
|
Bonnesen C, Nelander GM, Hansen BF, Jensen P, Krabbe JS, Jensen MB, Hegelund AC, Svendsen JE, Oleksiewicz MB. Synchronization in G0/G1 enhances the mitogenic response of cells overexpressing the human insulin receptor A isoform to insulin. Cell Biol Toxicol 2009; 26:293-307. [PMID: 19898946 PMCID: PMC2896650 DOI: 10.1007/s10565-009-9142-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Accepted: 10/22/2009] [Indexed: 01/01/2023]
Abstract
Evaluating mitogenic signaling specifically through the human insulin receptor (IR) is relevant for the preclinical safety assessment of developmental insulin analogs. It is known that overexpression of IR sensitizes cells to the mitogenic effects of insulin, but it is essentially unknown how mitogenic responses can be optimized to allow practical use of such recombinant cell lines for preclinical safety testing. We constitutively overexpressed the short isoform of the human insulin receptor (hIR-A, exon 11-negative) in L6 rat skeletal myoblasts. Because the mitogenic effect of growth factors such as insulin is expected to act in G0/G1, promoting S-phase entry, we developed a combined topoinhibition + serum deprivation strategy to explore the effect of G0/G1 synchronization as an independent parameter in the context of serum deprivation, the latter being routinely used to reduce background in mitogenicity assays. G0/G1 synchronization significantly improved the mitogenic responses of L6-hIR cells to insulin, measured by 3H-thymidine incorporation. Comparison with the parental L6 cells using phospho-mitogen-activated protein kinase, phospho-AKT, as well as 3H-thymidine incorporation end points supported that the majority of the mitogenic effect of insulin in L6-hIR cells was mediated by the overexpressed hIR-A. Using the optimized L6-hIR assay, we found that the X-10 insulin analog was more mitogenic than native human insulin, supporting that X-10 exhibits increased mitogenic signaling through the hIR-A. In summary, this study provides the first demonstration that serum deprivation may not be sufficient, and G0/G1 synchronization may be required to obtain optimal responsiveness of hIR-overexpressing cell lines for preclinical safety testing.
Collapse
|