1
|
Monteiro M, Zhang X, Yee D. Insulin promotes growth in breast cancer cells through the type I IGF receptor in insulin receptor deficient cells. Exp Cell Res 2024; 434:113862. [PMID: 38036052 PMCID: PMC10842809 DOI: 10.1016/j.yexcr.2023.113862] [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: 09/22/2023] [Revised: 11/14/2023] [Accepted: 11/21/2023] [Indexed: 12/02/2023]
Abstract
Breast cancer is the most common cancer in women. The upregulation of insulin-like growth factor (IGF) system observed in certain types of breast cancers was linked to growth, metastasis, and survival resulting in multiple strategies designed to target the type I IGF receptor (IGF-1R) in breast cancer. These attempts failed to prove beneficial and it has been suggested that insulin receptor (IR) could also play an important role in breast cancer biology. To better understand the IR's role in breast cancer cells, the receptor was deleted from MCF-7L cells using CRISPR technology, and fluorescence-assisted cell sorting was used to obtain clone 35 (CL35). It was found that CL35 activated signaling pathways upon insulin stimulation despite the absence of IR expression. We hypothesized that CL35 used a surrogate receptor for sustained growth and development. IGF-1R was able to activate insulin signaling and growth in CL35. Thus, insulin may play a central role in regulating breast cancer growth due to its ability to activate all the receptors of the IGF family. These findings argue that dual targeting of IR and IGF-IR may be required to inhibit breast cancer growth.
Collapse
Affiliation(s)
- Marvis Monteiro
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA; Purdue University, Heine Pharmacy Building, 575 Stadium Mall Drive, West Lafayette, IN, 47907-2091, USA
| | - Xihong Zhang
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Douglas Yee
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
2
|
Mapping the dynamics of insulin-responsive pathways in the blood-brain barrier endothelium using time-series transcriptomics data. NPJ Syst Biol Appl 2022; 8:29. [PMID: 35974022 PMCID: PMC9381797 DOI: 10.1038/s41540-022-00235-8] [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: 12/08/2021] [Accepted: 06/14/2022] [Indexed: 01/11/2023] Open
Abstract
Critical functions of the blood-brain barrier (BBB), including cerebral blood flow, energy metabolism, and immunomodulation, are regulated by insulin signaling pathways. Therefore, endothelial insulin resistance could lead to BBB dysfunction, which is associated with neurodegenerative diseases such as Alzheimer's disease (AD). The current study aims to map the dynamics of insulin-responsive pathways in polarized human cerebral microvascular endothelial cell (hCMEC/D3) monolayers. RNA-Sequencing was performed on hCMEC/D3 monolayers with and without insulin treatment at various time points. The Short Time-series Expression Miner (STEM) method was used to identify gene clusters with distinct and representative expression patterns. Functional annotation and pathway analysis of genes from selected clusters were conducted using Webgestalt and Ingenuity Pathway Analysis (IPA) software. Quantitative expression differences of 16,570 genes between insulin-treated and control monolayers were determined at five-time points. The STEM software identified 12 significant clusters with 6880 genes that displayed distinct temporal patterns upon insulin exposure, and the clusters were further divided into three groups. Gene ontology (GO) enrichment analysis demonstrated that biological processes protecting BBB functions such as regulation of vascular development and actin cytoskeleton reorganization were upregulated after insulin treatment (Group 1 and 2). In contrast, GO pathways related to inflammation, such as response to interferon-gamma, were downregulated (Group 3). The IPA analyses further identified insulin-responsive cellular and molecular pathways that are associated with AD pathology. These findings unravel the dynamics of insulin action on the BBB endothelium and inform about downstream signaling cascades that are potentially disrupted due to brain insulin resistance prevalent in AD.
Collapse
|
3
|
Hulett NA, Scalzo RL, Reusch JEB. Glucose Uptake by Skeletal Muscle within the Contexts of Type 2 Diabetes and Exercise: An Integrated Approach. Nutrients 2022; 14:647. [PMID: 35277006 PMCID: PMC8839578 DOI: 10.3390/nu14030647] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 02/01/2023] Open
Abstract
Type 2 diabetes continues to negatively impact the health of millions. The inability to respond to insulin to clear blood glucose (insulin resistance) is a key pathogenic driver of the disease. Skeletal muscle is the primary tissue for maintaining glucose homeostasis through glucose uptake via insulin-dependent and -independent mechanisms. Skeletal muscle is also responsive to exercise-meditated glucose transport, and as such, exercise is a cornerstone for glucose management in people with type 2 diabetes. Skeletal muscle glucose uptake requires a concert of events. First, the glucose-rich blood must be transported to the skeletal muscle. Next, the glucose must traverse the endothelium, extracellular matrix, and skeletal muscle membrane. Lastly, intracellular metabolic processes must be activated to maintain the diffusion gradient to facilitate glucose transport into the cell. This review aims to examine the physiology at each of these steps in healthy individuals, analyze the dysregulation affecting these pathways associated with type 2 diabetes, and describe the mechanisms by which exercise acts to increase glucose uptake.
Collapse
Affiliation(s)
- Nicholas A. Hulett
- Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA; (N.A.H.); (R.L.S.)
| | - Rebecca L. Scalzo
- Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA; (N.A.H.); (R.L.S.)
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO 80045, USA
- Center for Women’s Health Research, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA
| | - Jane E. B. Reusch
- Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA; (N.A.H.); (R.L.S.)
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO 80045, USA
- Center for Women’s Health Research, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA
| |
Collapse
|
4
|
Love KM, Barrett EJ, Malin SK, Reusch JEB, Regensteiner JG, Liu Z. Diabetes pathogenesis and management: the endothelium comes of age. J Mol Cell Biol 2021; 13:500-512. [PMID: 33787922 PMCID: PMC8530521 DOI: 10.1093/jmcb/mjab024] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/10/2021] [Accepted: 02/25/2021] [Indexed: 12/03/2022] Open
Abstract
Endothelium, acting as a barrier, protects tissues against factors that provoke insulin resistance and type 2 diabetes and itself responds to the insult of insulin resistance inducers with altered function. Endothelial insulin resistance and vascular dysfunction occur early in the evolution of insulin resistance-related disease, can co-exist with and even contribute to the development of metabolic insulin resistance, and promote vascular complications in those affected. The impact of endothelial insulin resistance and vascular dysfunction varies depending on the blood vessel size and location, resulting in decreased arterial plasticity, increased atherosclerosis and vascular resistance, and decreased tissue perfusion. Women with insulin resistance and diabetes are disproportionately impacted by cardiovascular disease, likely related to differential sex-hormone endothelium effects. Thus, reducing endothelial insulin resistance and improving endothelial function in the conduit arteries may reduce atherosclerotic complications, in the resistance arteries lead to better blood pressure control, and in the microvasculature lead to less microvascular complications and more effective tissue perfusion. Multiple diabetes therapeutic modalities, including medications and exercise training, improve endothelial insulin action and vascular function. This action may delay the onset of type 2 diabetes and/or its complications, making the vascular endothelium an attractive therapeutic target for type 2 diabetes and potentially type 1 diabetes.
Collapse
MESH Headings
- Age Factors
- Cardiovascular Diseases/epidemiology
- Cardiovascular Diseases/ethnology
- Cardiovascular Diseases/metabolism
- Cardiovascular Diseases/physiopathology
- Comorbidity
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/epidemiology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/physiopathology
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/epidemiology
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/physiopathology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Exercise
- Female
- Humans
- Hypoglycemic Agents/pharmacology
- Hypoglycemic Agents/therapeutic use
- Insulin Resistance
- Male
- Racial Groups
- Risk Factors
- Sex Factors
Collapse
Affiliation(s)
- Kaitlin M Love
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Eugene J Barrett
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Steven K Malin
- Department of Kinesiology and Health, Rutgers University, New Brunswick, NJ, USA
- Division of Endocrinology, Metabolism and Nutrition, Rutgers University, New Brunswick, NJ, USA
- New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, NJ, USA
- Institute of Translational Medicine and Research, Rutgers University, New Brunswick, NJ, USA
| | - Jane E B Reusch
- Center for Women’s Health Research, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, USA
| | - Judith G Regensteiner
- Center for Women’s Health Research, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Zhenqi Liu
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908, USA
| |
Collapse
|
5
|
Walker AMN, Warmke N, Mercer B, Watt NT, Mughal R, Smith J, Galloway S, Haywood NJ, Soomro T, Griffin KJ, Wheatcroft SB, Yuldasheva NY, Beech DJ, Carmeliet P, Kearney MT, Cubbon RM. Endothelial Insulin Receptors Promote VEGF-A Signaling via ERK1/2 and Sprouting Angiogenesis. Endocrinology 2021; 162:bqab104. [PMID: 34037749 PMCID: PMC8223729 DOI: 10.1210/endocr/bqab104] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Indexed: 02/08/2023]
Abstract
Endothelial insulin receptors (Insr) promote sprouting angiogenesis, although the underpinning cellular and molecular mechanisms are unknown. Comparing mice with whole-body insulin receptor haploinsufficiency (Insr+/-) against littermate controls, we found impaired limb perfusion and muscle capillary density after inducing hind-limb ischemia; this was in spite of increased expression of the proangiogenic growth factor Vegfa. Insr+/- neonatal retinas exhibited reduced tip cell number and branching complexity during developmental angiogenesis, which was also found in separate studies of mice with endothelium-restricted Insr haploinsufficiency. Functional responses to vascular endothelial growth factor A (VEGF-A), including in vitro angiogenesis, were also impaired in aortic rings and pulmonary endothelial cells from Insr+/- mice. Human umbilical vein endothelial cells with shRNA-mediated knockdown of Insr also demonstrated impaired functional angiogenic responses to VEGF-A. VEGF-A signaling to Akt and endothelial nitric oxide synthase was intact, but downstream signaling to extracellular signal-reduced kinase 1/2 (ERK1/2) was impaired, as was VEGF receptor-2 (VEGFR-2) internalization, which is required specifically for signaling to ERK1/2. Hence, endothelial insulin receptors facilitate the functional response to VEGF-A during angiogenic sprouting and are required for appropriate signal transduction from VEGFR-2 to ERK1/2.
Collapse
Affiliation(s)
- Andrew M N Walker
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds LS2 9JT, UK
| | - Nele Warmke
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds LS2 9JT, UK
| | - Ben Mercer
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds LS2 9JT, UK
| | - Nicole T Watt
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds LS2 9JT, UK
| | - Romana Mughal
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds LS2 9JT, UK
| | - Jessica Smith
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds LS2 9JT, UK
| | - Stacey Galloway
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds LS2 9JT, UK
| | - Natalie J Haywood
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds LS2 9JT, UK
| | - Taha Soomro
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds LS2 9JT, UK
- Imperial College Ophthalmology Research Group, Western Eye Hospital, London NW1 5QH, UK
| | - Kathryn J Griffin
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds LS2 9JT, UK
| | - Stephen B Wheatcroft
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds LS2 9JT, UK
| | - Nadira Y Yuldasheva
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds LS2 9JT, UK
| | - David J Beech
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds LS2 9JT, UK
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Department of Oncology, University of Leuven, Leuven 3000, Belgium
| | - Mark T Kearney
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds LS2 9JT, UK
| | - Richard M Cubbon
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
6
|
Abstract
CONTEXT Obesity and type 2 diabetes are associated with chronic hyperinsulinemia, elevated plasma levels of dipeptidyl peptidase-4 (DPP4), and a pro-atherosclerotic milieu. EVIDENCE ACQUISITION PubMed search of the term "insulin and atherosclerosis," "hyperinsulinemia," "atherosclerosis," or "cardiovascular outcomes" cross-referenced with "DPP4." Relevant research and review articles were reviewed. EVIDENCE SYNTHESIS Hyperinsulinemia in the setting of insulin resistance promotes vascular inflammation, vascular smooth muscle cell growth, pathological cholesterol profile, hypertension, and recruitment of immune cells to the endothelium, all contributing to atherosclerosis. DPP4 has pleiotropic functions and its activity is elevated in obese humans. DPP4 mirrors hyperinsulinemia's atherogenic actions in the insulin resistant state, and genetic deletion of DPP4 protects rodents from developing insulin resistance and improves cardiovascular outcomes. DPP4 inhibition in pro-atherosclerotic preclinical models results in reduced inflammation and oxidative stress, improved endothelial function, and decreased atherosclerosis. Increased incretin levels may have contributed to but do not completely account for these benefits. Small clinical studies with DPP4 inhibitors demonstrate reduced carotid intimal thickening, improved endothelial function, and reduced arterial stiffness. To date, this has not been translated to cardiovascular risk reduction for individuals with type 2 diabetes with prior or exaggerated risk of cardiovascular disease. CONCLUSION DPP4 may represent a key link between central obesity, insulin resistance, and atherosclerosis. The gaps in knowledge in DPP4 function and discrepancy in cardiovascular outcomes observed in preclinical and large-scale randomized controlled studies with DPP4 inhibitors warrant additional research.
Collapse
Affiliation(s)
- Kaitlin M Love
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA, USA
| | - Zhenqi Liu
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA, USA
- Correspondence: Zhenqi Liu, M.D., University of Virginia Health System, Charlottesville, VA, USA.
| |
Collapse
|
7
|
Oghbaei H, Fattahi A, Hamidian G, Sadigh-Eteghad S, Ziaee M, Mahmoudi J. A closer look at the role of insulin for the regulation of male reproductive function. Gen Comp Endocrinol 2021; 300:113643. [PMID: 33017586 DOI: 10.1016/j.ygcen.2020.113643] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/16/2020] [Accepted: 09/28/2020] [Indexed: 11/25/2022]
Abstract
While insulin demonstrates to have a considerable influence on the reproductive system, there are various unanswered questions regarding its precise sites, mechanisms of action, and roles for the developing and functioning of the adult male reproductive system. Apart from its effects on glucose level, insulin has an important role in the reproductive system directly by binding on insulin and IGF receptors in the brain and testis. To date, however, the effect of insulin or its alterations on blood-testis-barrier, as an important regulator of normal spermatogenesis and fertility, has not yet been studied. This review aimed to focus on the experimental and clinical studies to describe mechanisms by which insulin affects the hypothalamic-pituitary-gonadal (HPG) axis, testicular cells, spermatozoa, and sexual behavior. Moreover, we discussed the mechanism and impact of insulin changes in type 1 (insulin deficiency along with persisted or even increased sensitivity) and 2 (insulin resistance along with increased insulin level at the early stages of disease) diabetes and obesity on the male reproductive tract.
Collapse
Affiliation(s)
- Hajar Oghbaei
- Department of Physiology, Tabriz University of Medical Sciences, Tabriz, Iran; Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Fattahi
- Department of Reproductive Biology, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - GholamReza Hamidian
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mojtaba Ziaee
- Medicinal Plants Research Center, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
8
|
Muniyappa R, Chen H, Montagnani M, Sherman A, Quon MJ. Endothelial dysfunction due to selective insulin resistance in vascular endothelium: insights from mechanistic modeling. Am J Physiol Endocrinol Metab 2020; 319:E629-E646. [PMID: 32776829 PMCID: PMC7642854 DOI: 10.1152/ajpendo.00247.2020] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Previously, we have used mathematical modeling to gain mechanistic insights into insulin-stimulated glucose uptake. Phosphatidylinositol 3-kinase (PI3K)-dependent insulin signaling required for metabolic actions of insulin also regulates endothelium-dependent production of the vasodilator nitric oxide (NO). Vasodilation increases blood flow that augments direct metabolic actions of insulin in skeletal muscle. This is counterbalanced by mitogen-activated protein kinase (MAPK)-dependent insulin signaling in endothelium that promotes secretion of the vasoconstrictor endothelin-1 (ET-1). In the present study, we extended our model of metabolic insulin signaling into a dynamic model of insulin signaling in vascular endothelium that explicitly represents opposing PI3K/NO and MAPK/ET-1 pathways. Novel NO and ET-1 subsystems were developed using published and new experimental data to generate model structures/parameters. The signal-response relationships of our model with respect to insulin-stimulated NO production, ET-1 secretion, and resultant vascular tone, agree with published experimental data, independent of those used for model development. Simulations of pathological stimuli directly impairing only insulin-stimulated PI3K/Akt activity predict altered dynamics of NO and ET-1 consistent with endothelial dysfunction in insulin-resistant states. Indeed, modeling pathway-selective impairment of PI3K/Akt pathways consistent with insulin resistance caused by glucotoxicity, lipotoxicity, or inflammation predict diminished NO production and increased ET-1 secretion characteristic of diabetes and endothelial dysfunction. We conclude that our mathematical model of insulin signaling in vascular endothelium supports the hypothesis that pathway-selective insulin resistance accounts, in part, for relationships between insulin resistance and endothelial dysfunction. This may be relevant for developing novel approaches for the treatment of diabetes and its cardiovascular complications.
Collapse
Affiliation(s)
- Ranganath Muniyappa
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | - Hui Chen
- Clinical and Integrative Diabetes and Obesity Integrated Review Group, Center for Scientific Review, National Institutes of Health, Bethesda, Maryland
| | - Monica Montagnani
- Department of Biomedical Sciences and Human Oncology, Pharmacology Section, Medical School, University of Bari "Aldo Moro", Bari, Italy
| | - Arthur Sherman
- Laboratory of Biological Modeling, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | - Michael J Quon
- Laboratory of Biological Modeling, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
- Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
9
|
Kondash ME, Ananthakumar A, Khodabukus A, Bursac N, Truskey GA. Glucose Uptake and Insulin Response in Tissue-engineered Human Skeletal Muscle. Tissue Eng Regen Med 2020; 17:801-813. [PMID: 32200516 DOI: 10.1007/s13770-020-00242-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 01/19/2020] [Accepted: 01/21/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Tissue-engineered muscles ("myobundles") offer a promising platform for developing a human in vitro model of healthy and diseased muscle for drug development and testing. Compared to traditional monolayer cultures, myobundles better model the three-dimensional structure of native skeletal muscle and are amenable to diverse functional measures to monitor the muscle health and drug response. Characterizing the metabolic function of human myobundles is of particular interest to enable their utilization in mechanistic studies of human metabolic diseases, identification of related drug targets, and systematic studies of drug safety and efficacy. METHODS To this end, we studied glucose uptake and insulin responsiveness in human tissue-engineered skeletal muscle myobundles in the basal state and in response to drug treatments. RESULTS In the human skeletal muscle myobundle system, insulin stimulates a 50% increase in 2-deoxyglucose (2-DG) uptake with a compiled EC50 of 0.27 ± 0.03 nM. Treatment of myobundles with 400 µM metformin increased basal 2-DG uptake 1.7-fold and caused a significant drop in twitch and tetanus contractile force along with decreased fatigue resistance. Treatment with the histone deacetylase inhibitor 4-phenylbutyrate (4-PBA) increased the magnitude of insulin response from a 1.2-fold increase in glucose uptake in the untreated state to a 1.4-fold increase after 4-PBA treatment. 4-PBA treated myobundles also exhibited increased fatigue resistance and increased twitch half-relaxation time. CONCLUSION Although tissue-engineered human myobundles exhibit a modest increase in glucose uptake in response to insulin, they recapitulate key features of in vivo insulin sensitivity and exhibit relevant drug-mediated perturbations in contractile function and glucose metabolism.
Collapse
Affiliation(s)
- Megan E Kondash
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | | | - Alastair Khodabukus
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA.
| |
Collapse
|
10
|
Wang N, Tan AWK, Jahn LA, Hartline L, Patrie JT, Lin S, Barrett EJ, Aylor KW, Liu Z. Vasodilatory Actions of Glucagon-Like Peptide 1 Are Preserved in Skeletal and Cardiac Muscle Microvasculature but Not in Conduit Artery in Obese Humans With Vascular Insulin Resistance. Diabetes Care 2020; 43:634-642. [PMID: 31888883 PMCID: PMC7035589 DOI: 10.2337/dc19-1465] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/02/2019] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Obesity is associated with microvascular insulin resistance, which is characterized by impaired insulin-mediated microvascular recruitment. Glucagon-like peptide 1 (GLP-1) recruits skeletal and cardiac muscle microvasculature, and this action is preserved in insulin-resistant rodents. We aimed to examine whether GLP-1 recruits microvasculature and improves the action of insulin in obese humans. RESEARCH DESIGN AND METHODS Fifteen obese adults received intravenous infusion of either saline or GLP-1 (1.2 pmol/kg/min) for 150 min with or without a euglycemic insulin clamp (1 mU/kg/min) superimposed over the last 120 min. Skeletal and cardiac muscle microvascular blood volume (MBV), flow velocity and blood flow, brachial artery diameter and blood flow, and pulse wave velocity (PWV) were determined. RESULTS Insulin failed to change MBV or flow in either skeletal or cardiac muscle, confirming the presence of microvascular insulin resistance. GLP-1 infusion alone increased MBV by ∼30% and ∼40% in skeletal and cardiac muscle, respectively, with no change in flow velocity, leading to a significant increase in microvascular blood flow in both skeletal and cardiac muscle. Superimposition of insulin to GLP-1 infusion did not further increase MBV or flow in either skeletal or cardiac muscle but raised the steady-state glucose infusion rate by ∼20%. Insulin, GLP-1, and GLP-1 + insulin infusion did not alter brachial artery diameter and blood flow or PWV. The vasodilatory actions of GLP-1 are preserved in both skeletal and cardiac muscle microvasculature, which may contribute to improving metabolic insulin responses and cardiovascular outcomes. CONCLUSIONS In obese humans with microvascular insulin resistance, GLP-1's vasodilatory actions are preserved in both skeletal and cardiac muscle microvasculature, which may contribute to improving metabolic insulin responses and cardiovascular outcomes.
Collapse
Affiliation(s)
- Nasui Wang
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA
- Department of Endocrinology and Metabolism, Shantou University First Hospital, Guangdong, China
| | - Alvin W K Tan
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore
| | - Linda A Jahn
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA
| | - Lee Hartline
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA
| | - James T Patrie
- Department of Public Health Sciences, University of Virginia Health System, Charlottesville, VA
| | - Shaoda Lin
- Department of Endocrinology and Metabolism, Shantou University First Hospital, Guangdong, China
| | - Eugene J Barrett
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA
| | - Kevin W Aylor
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA
| | - Zhenqi Liu
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA
| |
Collapse
|
11
|
Sheng X, Yao K, Shao A, Tu S, Zhang X, Chen T, Yao D. The Role of Insulin Glargine and Human Insulin in the Regulation of Thyroid Proliferation Through Mitogenic Signaling. Front Endocrinol (Lausanne) 2019; 10:594. [PMID: 31555212 PMCID: PMC6723759 DOI: 10.3389/fendo.2019.00594] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 08/13/2019] [Indexed: 12/29/2022] Open
Abstract
Our aim was to investigate whether human insulin (HI) or insulin glargine treatment could promote the proliferation of thyroid cells and determine the association between type 2 diabetes and thyroid disease. Rats were treated with different doses of HI and insulin glargine. Plasma glucose and the phosphorylation levels of the insulin receptor (IR), insulin-like growth factor 1 receptor (IGF-1R), protein kinase B (Akt), and extracellular signal-regulated kinase 1/2 (ERK1/2) were measured. A total of 105 rats were randomly assigned to three groups as follows: control group, HI group, and glargine group. Both drugs promoted the phosphorylation of IR, Akt, and ERK1/2 in a dose-dependent manner (p < 0.05), and the effect of glargine persisted for longer period. Treatment with ultra-therapeutic doses of HI or glargine (p < 0.05) increased the expression of Ki-67 in thyroid cells. The results demonstrated that therapeutic doses of glargine have a longer-lasting hypoglycemic control than HI. Based on the results, HI or glargine did not stimulate thyroid cell proliferation at therapeutic doses, but high doses did.
Collapse
Affiliation(s)
- Xiaoli Sheng
- Department of Obstetrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kannan Yao
- The Second Central Laboratory, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Sheng Tu
- Department of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xinxia Zhang
- Department of Geriatrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ting Chen
- Department of Ultrasonography, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Dingguo Yao
- Department of Endocrinology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
12
|
Ramos DM, Abdulmalik S, Arul MR, Rudraiah S, Laurencin CT, Mazzocca AD, Kumbar SG. Insulin immobilized PCL-cellulose acetate micro-nanostructured fibrous scaffolds for tendon tissue engineering. POLYM ADVAN TECHNOL 2019; 30:1205-1215. [PMID: 30956516 PMCID: PMC6448803 DOI: 10.1002/pat.4553] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 01/03/2019] [Indexed: 12/28/2022]
Abstract
Use of growth factors as biochemical molecules to elicit cellular differentiation is a common strategy in tissue engineering. However, limitations associated with growth factors, such as short half-life, high effective physiological doses, and high costs, have prompted the search for growth factor alternatives, such as growth factor mimics and other proteins. This work explores the use of insulin protein as a biochemical factor to aid in tendon healing and differentiation of cells on a biomimetic electrospun micro-nanostructured scaffold. Dose response studies were conducted using human mesenchymal stem cells (MSCs) in basal media supplemented with varied insulin concentrations. A dose of 100-ng/mL insulin showed increased expression of tendon markers. Synthetic-natural blends of various ratios of polycaprolactone (PCL) and cellulose acetate (CA) were used to fabricate micro-nanofibers to balance physicochemical properties of the scaffolds in terms of mechanical strength, hydrophilicity, and insulin delivery. A 75:25 ratio of PCL:CA was found to be optimal in promoting cellular attachment and insulin immobilization. Insulin insulin deliveryimmobilized fiber matrices also showed increased expression of tendon phenotypic markers by MSCs similar to findings with insulin supplemented media, indicating preservation of insulin bioactivity. Insulin functionalized scaffolds may have potential applications in tendon healing and regeneration.
Collapse
Affiliation(s)
- Daisy M. Ramos
- Department of Materials Science and Engineering, University of Connecticut, Storrs, Connecticut
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, Connecticut
| | - Sama Abdulmalik
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, Connecticut
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut
| | - Michael R. Arul
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, Connecticut
| | - Swetha Rudraiah
- Department of Pharmaceutical Sciences, University of Saint Joseph, Hartford, Connecticut
| | - Cato T. Laurencin
- Department of Materials Science and Engineering, University of Connecticut, Storrs, Connecticut
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, Connecticut
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut
| | - Augustus D. Mazzocca
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, Connecticut
| | - Sangamesh G. Kumbar
- Department of Materials Science and Engineering, University of Connecticut, Storrs, Connecticut
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, Connecticut
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut
| |
Collapse
|
13
|
Mughal RS, Bridge K, Buza I, Slaaby R, Worm J, Klitgaard-Povlsen G, Hvid H, Schiødt M, Cubbon R, Yuldasheva N, Skromna A, Makava N, Skytte-Olsen G, Kearney MT. Effects of obesity on insulin: insulin-like growth factor 1 hybrid receptor expression and Akt phosphorylation in conduit and resistance arteries. Diab Vasc Dis Res 2019; 16:160-170. [PMID: 30295509 PMCID: PMC6484231 DOI: 10.1177/1479164118802550] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Insulin and insulin-like growth factor-1 stimulate specific responses in arteries, which may be disrupted by diet-induced obesity. We examined (1) temporal effects of high-fat diet compared to low-fat diet in mice on insulin receptor, insulin-like growth factor-1 receptor, insulin receptor/insulin-like growth factor-1 receptor hybrid receptor expression and insulin/insulin-like growth factor-1-mediated Akt phosphorylation in aorta; and (2) effects of high-fat diet on insulin and insulin-like growth factor-1-mediated Akt phosphorylation and vascular tone in resistance arteries. Medium-term high-fat diet (5 weeks) decreased insulin-like growth factor-1 receptor expression and increased hybrid expression (~30%) only. After long-term (16 weeks) high-fat diet, insulin receptor expression was reduced by ~30%, insulin-like growth factor-1 receptor expression decreased a further ~40% and hybrid expression increased a further ~60%. Independent correlates of hybrid receptor expression were high-fat diet, duration of high-fat diet and plasma insulin-like growth factor-1 (all p < 0.05). In aorta, insulin was a more potent activator of Akt than insulin-like growth factor-1, whereas in resistance arteries, insulin-like growth factor-1 was more potent than insulin. High-fat diet blunted insulin-mediated vasorelaxation ( p < 0.01) but had no effect on insulin-like growth factor-1-mediated vasorelaxation in resistance arteries. Our findings support the possibility that hybrid receptor level is influenced by nutritional and metabolic cues. Moreover, vessel-dependent effects of insulin and insulin-like growth factor-1 on vascular tone and Akt activation may have implications in treating obesity-related vascular disease.
Collapse
MESH Headings
- Animals
- Antigens, CD/metabolism
- Aorta/drug effects
- Aorta/enzymology
- Cells, Cultured
- Diet, Fat-Restricted
- Diet, High-Fat
- Disease Models, Animal
- Enzyme Activation
- Human Umbilical Vein Endothelial Cells/drug effects
- Human Umbilical Vein Endothelial Cells/enzymology
- Humans
- Insulin/pharmacology
- Insulin-Like Growth Factor I/pharmacology
- Male
- Mesenteric Arteries/drug effects
- Mesenteric Arteries/enzymology
- Mesenteric Arteries/physiopathology
- Mice, Inbred C57BL
- Obesity/blood
- Obesity/enzymology
- Obesity/physiopathology
- Phosphorylation
- Proto-Oncogene Proteins c-akt/metabolism
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/metabolism
- Receptor, Insulin/metabolism
- Receptors, Somatomedin/metabolism
- Signal Transduction/drug effects
- Vascular Resistance/drug effects
- Vasodilation/drug effects
Collapse
Affiliation(s)
- Romana S Mughal
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), School of Medicine, University of Leeds, Leeds, UK
| | - Katherine Bridge
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), School of Medicine, University of Leeds, Leeds, UK
| | - Irma Buza
- Global Research, Novo Nordisk A/S, Malov, Denmark
| | - Rita Slaaby
- Global Research, Novo Nordisk A/S, Malov, Denmark
| | - Jesper Worm
- Global Research, Novo Nordisk A/S, Malov, Denmark
| | | | - Henning Hvid
- Global Research, Novo Nordisk A/S, Malov, Denmark
| | | | - Richard Cubbon
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), School of Medicine, University of Leeds, Leeds, UK
| | - Nadira Yuldasheva
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), School of Medicine, University of Leeds, Leeds, UK
| | - Anna Skromna
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), School of Medicine, University of Leeds, Leeds, UK
| | - Natallia Makava
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), School of Medicine, University of Leeds, Leeds, UK
| | | | - Mark T Kearney
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), School of Medicine, University of Leeds, Leeds, UK
- Mark T Kearney, Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), School of Medicine, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
14
|
Soliman GF, Rashed LA, Morsi H, Ibrahim W, Abdallah H, Bastawy N, Abdel Maksoud OM. Interrelation of liver vascularity to non-alcoholic fatty liver through a comparative study of the vasodilator effect of carvedilol or nicorandil in rats. Life Sci 2019; 222:175-182. [PMID: 30826497 DOI: 10.1016/j.lfs.2019.02.057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 02/20/2019] [Accepted: 02/27/2019] [Indexed: 12/30/2022]
Abstract
AIM An experimental study of the effect of two vasodilators, carvedilol (B blocker with alpha-antagonist) and nicorandil (NO donor) on nonalcoholic fatty liver (NAFLD) induced by hypercholesterolemia and fatty diet in rats through studying the possible anti-inflammatory and antioxidant mechanisms. MAIN METHODS The rats were divided into 4 groups (6 rats each): The first (negative control group). The second, third and fourth groups were fed with cholesterol and fat- enriched diet for one month that stopped and continued on the standard diet for another month without treatment in the second group but treated with carvedilol and nicorandil in the third and fourth group respectively. KEY FINDINGS They revealed that both improved NAFLD especially nicorandil treated proved by the reduction of liver enzymes (AST, ALT), the fatty infiltration determined histologically and biochemically (decrease liver triglycerides). This may be due to either being antioxidants (reduced malondialdehyde and elevated reduced glutathione) or anti-inflammatory (decreased of TNF-α) together with the reduction of insulin resistance and adiponectin elevation or gene expression (increased liver NF-κB and decreased eNOS expression) and finally maybe by their obvious effect on improvements of lipid parameters. SIGNIFICANCE Carvedilol and nicorandil improved NAFLD through the interrelationship between inflammatory cytokines, antioxidants and insulin resistance.
Collapse
Affiliation(s)
- Ghada Farouk Soliman
- Department of Medical Pharmacology, Faculty of Medicine, Cairo University, Egypt.
| | - Laila Ahmed Rashed
- Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Egypt
| | - Heba Morsi
- Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Egypt
| | - Walaa Ibrahim
- Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Egypt
| | - Hanan Abdallah
- Department of Medical Histology, Faculty of Medicine, Cairo University, Egypt
| | - Nermeen Bastawy
- Department of Medical Physiology, Faculty of Medicine, Cairo University, Egypt
| | | |
Collapse
|
15
|
Zhang X, Sheng X, Miao T, Yao K, Yao D. Effect of insulin on thyroid cell proliferation, tumor cell migration, and potentially related mechanisms. Endocr Res 2019; 44:55-70. [PMID: 30260725 DOI: 10.1080/07435800.2018.1522641] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Diabetes has recently been identified as a risk factor for a variety of cancers, possibly due to hyperinsulinemia or exogenous insulin use. Thyroid cancer is the most common endocrine malignancy, and its incidence has been exponentially increasing worldwide at an alarming rate. The aim of this study was to establish whether insulin use affects thyroid cancer development and progression, specifically cell proliferation and migration in vitro. METHODS In this study, we investigated the effects of the insulin agents most commonly used in the clinic, regular human insulin (HI) and insulin glargine (IG), on the proliferation and migration of thyroid cells. RESULTS Both HI and IG affected the thyroid cells in a dose-dependent manner and at high concentrations significantly promoted thyroid cell proliferation and tumor cell migration. The promoting effect might be elicited by activation of the insulin receptor and insulin-like growth factor-1 receptor and through the downstream Akt-signaling pathway, which inhibits the activity of the tumor-suppressor FoxO3a. In particular, MAPK-signaling cascades were activated in papillary thyroid carcinoma cell-1 cells but not in follicular rat thyroid-5 cells. CONCLUSION The in vitro evidence demonstrated that HI and IG can promote thyroid cell proliferation and tumor cell migration at supraphysiological concentrations, but the effect was not significant at low concentrations. Whether high-dose insulins could affect diabetic patients with thyroid cancer or undetected (pre)cancerous lesions needs further in vivo study. ABBREVIATIONS HI: human regular insulin; IG: insulin glargine; IR: insulin receptor; IGF-1R: insulin-like growth factor-1 receptor; Akt: protein kinase B (PKB); MAPK: mitogen-activated protein kinase; FoxO3a: the forkhead box-containing protein: class O 3a.
Collapse
Affiliation(s)
- Xinxia Zhang
- a Department of Geriatrics , The First Affiliated Hospital, College of Medicine, Zhejiang University , Hangzhou , Zhejiang , China
| | - Xiaoli Sheng
- b Department of Obstetrics , The First Affiliated Hospital, College of Medicine, Zhejiang University , Hangzhou , Zhejiang , China
| | - Tingru Miao
- c ECG Functional Department , Zhejiang Province People's Hospital , Hangzhou , Zhejiang , China
| | - Kannan Yao
- d The Second Central Laboratory , The First Affiliated Hospital of Zhejiang Chinese Medical University , Hangzhou , Zhejiang , China
| | - Dingguo Yao
- e Department of Endocrinology , The First Affiliated Hospital of Zhejiang Chinese Medical University , Hangzhou , Zhejiang , China
| |
Collapse
|
16
|
Muscle Insulin Resistance and the Inflamed Microvasculature: Fire from Within. Int J Mol Sci 2019; 20:ijms20030562. [PMID: 30699907 PMCID: PMC6387226 DOI: 10.3390/ijms20030562] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 01/21/2019] [Accepted: 01/22/2019] [Indexed: 12/18/2022] Open
Abstract
Insulin is a vascular hormone and regulates vascular tone and reactivity. Muscle is a major insulin target that is responsible for the majority of insulin-stimulated glucose use. Evidence confirms that muscle microvasculature is an important insulin action site and critically regulates insulin delivery to muscle and action on myocytes, thereby affecting insulin-mediated glucose disposal. Insulin via activation of its signaling cascade in the endothelial cells increases muscle microvascular perfusion, which leads to an expansion of the endothelial exchange surface area. Insulin’s microvascular actions closely couple with its metabolic actions in muscle and blockade of insulin-mediated microvascular perfusion reduces insulin-stimulated muscle glucose disposal. Type 2 diabetes is associated with chronic low-grade inflammation, which engenders both metabolic and microvascular insulin resistance through endocrine, autocrine and paracrine actions of multiple pro-inflammatory factors. Here, we review the crucial role of muscle microvasculature in the regulation of insulin action in muscle and how inflammation in the muscle microvasculature affects insulin’s microvascular actions as well as metabolic actions. We propose that microvascular insulin resistance induced by inflammation is an early event in the development of metabolic insulin resistance and eventually type 2 diabetes and its related cardiovascular complications, and thus is a potential therapeutic target for the prevention or treatment of obesity and diabetes.
Collapse
|
17
|
Maffei P, Dassie F, Wennberg A, Parolin M, Vettor R. The Endothelium in Acromegaly. Front Endocrinol (Lausanne) 2019; 10:437. [PMID: 31396153 PMCID: PMC6667653 DOI: 10.3389/fendo.2019.00437] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 06/18/2019] [Indexed: 12/12/2022] Open
Abstract
Growth hormone (GH) and insulin like growth factor-1 (IGF-1) excess induce well-known deleterious effects on the cardiovascular system, especially after long-term exposition. Acromegaly, a condition of chronic GH and IGF-1 hypersecretion, is frequently associated to cardiovascular complications, although recent studies have shown a reduction in the prevalence of these comorbidities in well-controlled patients and a mortality risk similar to normal aging population. Many factors could contribute to the increased cardiovascular risk of acromegaly patients. Among these factors, the endothelium plays a key role in the pathogenesis of atherosclerotic plaques and could be considered an early marker of atherosclerosis and cardiovascular dysfunction. In this review we examined the relationship between GH/IGF-1 excess and the endothelium, from basic studies to clinical evidence. Many studies involving various arterial districts (microvascular arteries of retina, kidney and brain, and major vessels as carotid and aorta) showed that GH/IGF-1 excess promotes endothelial dysfunction via several different mechanisms. Increased endothelial proliferation, dysfunction of endothelial progenitor cells, increased oxidative stress, and compromised oxidative defenses are the main factors that are associated with endothelial dysfunction. In the general population, these alterations are associated with the development of atherosclerosis with an increased incidence of coronary artery disease and cerebrovascular complications. However, in acromegaly this is still a debated issue, despite the presence of many pro-atherogenic factors and comorbidities, such as hypertension, diabetes, sleep apnoea, and metabolic syndrome. Preclinical markers of atherosclerosis as arterial intima media thickness, pulse wave velocity and flow mediated dilation seem to be impaired in acromegaly and partly mediated by the endothelium dysfunction. In conclusion, the pathophysiology of endothelial dysfunction in the condition of GH and IGF-1 excess remains a crucial area of investigation to fully dissect the association of acromegaly with cardiovascular disease complications.
Collapse
Affiliation(s)
- Pietro Maffei
- Clinica Medica 3, Department of Medicine (DIMED), Padua University Hospital, Padua, Italy
- *Correspondence: Pietro Maffei
| | - Francesca Dassie
- Clinica Medica 3, Department of Medicine (DIMED), Padua University Hospital, Padua, Italy
| | - Alexandra Wennberg
- Clinica Neurologica, Department of Neurosciences (DNS), Padua University Hospital, Padua, Italy
| | - Matteo Parolin
- Clinica Medica 3, Department of Medicine (DIMED), Padua University Hospital, Padua, Italy
| | - Roberto Vettor
- Clinica Medica 3, Department of Medicine (DIMED), Padua University Hospital, Padua, Italy
| |
Collapse
|
18
|
Ntoumou E, Tzetis M, Braoudaki M, Lambrou G, Poulou M, Malizos K, Stefanou N, Anastasopoulou L, Tsezou A. Serum microRNA array analysis identifies miR-140-3p, miR-33b-3p and miR-671-3p as potential osteoarthritis biomarkers involved in metabolic processes. Clin Epigenetics 2017; 9:127. [PMID: 29255496 PMCID: PMC5728069 DOI: 10.1186/s13148-017-0428-1] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 11/29/2017] [Indexed: 02/06/2023] Open
Abstract
Background MicroRNAs (miRNAs) in circulation have emerged as promising biomarkers. In this study, we aimed to identify a circulating miRNA signature for osteoarthritis (OA) patients and in combination with bioinformatics analysis to evaluate the utility of selected differentially expressed miRNAs in the serum as potential OA biomarkers. Methods Serum samples were collected from 12 primary OA patients, and 12 healthy individuals were screened using the Agilent Human miRNA Microarray platform interrogating 2549 miRNAs. Receiver Operating Characteristic (ROC) curves were constructed to evaluate the diagnostic performance of the deregulated miRNAs. Expression levels of selected miRNAs were validated by quantitative real-time PCR (qRT-PCR) in all serum and in articular cartilage samples from OA patients (n = 12) and healthy individuals (n = 7). Bioinformatics analysis was used to investigate the involved pathways and target genes for the above miRNAs. Results We identified 279 differentially expressed miRNAs in the serum of OA patients compared to controls. Two hundred and five miRNAs (73.5%) were upregulated and 74 (26.5%) downregulated. ROC analysis revealed that 77 miRNAs had area under the curve (AUC) > 0.8 and p < 0.05. Bioinformatics analysis in the 77 miRNAs revealed that their target genes were involved in multiple signaling pathways associated with OA, among which FoxO, mTOR, Wnt, pI3K/akt, TGF-β signaling pathways, ECM-receptor interaction, and fatty acid biosynthesis. qRT-PCR validation in seven selected out of the 77 miRNAs revealed 3 significantly downregulated miRNAs (hsa-miR-33b-3p, hsa-miR-671-3p, and hsa-miR-140-3p) in the serum of OA patients, which were in silico predicted to be enriched in pathways involved in metabolic processes. Target-gene analysis of hsa-miR-140-3p, hsa-miR-33b-3p, and hsa-miR-671-3p revealed that InsR and IGFR1 were common targets of all three miRNAs, highlighting their involvement in regulation of metabolic processes that contribute to OA pathology. Hsa-miR-140-3p and hsa-miR-671-3p expression levels were consistently downregulated in articular cartilage of OA patients compared to healthy individuals. Conclusions A serum miRNA signature was established for the first time using high density resolution miR-arrays in OA patients. We identified a three-miRNA signature, hsa-miR-140-3p, hsa-miR-671-3p, and hsa-miR-33b-3p, in the serum of OA patients, predicted to regulate metabolic processes, which could serve as a potential biomarker for the evaluation of OA risk and progression. Electronic supplementary material The online version of this article (10.1186/s13148-017-0428-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- E Ntoumou
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, Biopolis, University of Thessaly, 41500 Larissa, Greece
| | - M Tzetis
- Laboratory of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - M Braoudaki
- Laboratory of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens,, Athens, Greece.,University Research Institute for the Study and Treatment of Childhood Disease and Malignant Diseases, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - G Lambrou
- First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens,, Athens, Greece.,University Research Institute for the Study and Treatment of Childhood Disease and Malignant Diseases, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - M Poulou
- Laboratory of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - K Malizos
- Department of Orthopaedics, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - N Stefanou
- Department of Orthopaedics, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - L Anastasopoulou
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, Biopolis, University of Thessaly, 41500 Larissa, Greece
| | - A Tsezou
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, Biopolis, University of Thessaly, 41500 Larissa, Greece.,Department of Biology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| |
Collapse
|
19
|
Pasarín M, Abraldes JG, Liguori E, Kok B, La Mura V. Intrahepatic vascular changes in non-alcoholic fatty liver disease: Potential role of insulin-resistance and endothelial dysfunction. World J Gastroenterol 2017; 23:6777-6787. [PMID: 29085222 PMCID: PMC5645612 DOI: 10.3748/wjg.v23.i37.6777] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 09/06/2017] [Accepted: 09/19/2017] [Indexed: 02/06/2023] Open
Abstract
Metabolic syndrome is a cluster of several clinical conditions characterized by insulin-resistance and high cardiovascular risk. Non-alcoholic fatty liver disease is the liver expression of the metabolic syndrome, and insulin resistance can be a frequent comorbidity in several chronic liver diseases, in particular hepatitis C virus infection and/or cirrhosis. Several studies have demonstrated that insulin action is not only relevant for glucose control, but also for vascular homeostasis. Insulin regulates nitric oxide production, which mediates to a large degree the vasodilating, anti-inflammatory and antithrombotic properties of a healthy endothelium, guaranteeing organ perfusion. The effects of insulin on the liver microvasculature and the effects of IR on sinusoidal endothelial cells have been studied in animal models of non-alcoholic fatty liver disease. The hypotheses derived from these studies and the potential translation of these results into humans are critically discussed in this review.
Collapse
Affiliation(s)
- Marcos Pasarín
- Hepatic Hemodynamic Laboratory, Liver Unit, Hospital Clinic, IDIBAPS (Institut d’Investigacions Biomèdiques August Pi i Sunyer), University of Barcelona, 08036 Barcelona, Spain
| | - Juan G Abraldes
- Cirrhosis Care Clinic, Division of Gastroenterology (Liver Unit), CEGIIR, University of Alberta, AB T6G 2R3 Edmonton, Canada
| | - Eleonora Liguori
- Internal Medicine, IRCCS San Donato, Department of Biomedical Sciences for Health, University of Milan, 20097 San Donato Milanese, Italy
| | - Beverley Kok
- Cirrhosis Care Clinic, Division of Gastroenterology (Liver Unit), CEGIIR, University of Alberta, AB T6G 2R3 Edmonton, Canada
| | - Vincenzo La Mura
- Internal Medicine, IRCCS San Donato, Department of Biomedical Sciences for Health, University of Milan, 20097 San Donato Milanese, Italy
| |
Collapse
|
20
|
Endothelial insulin receptors differentially control insulin signaling kinetics in peripheral tissues and brain of mice. Proc Natl Acad Sci U S A 2017; 114:E8478-E8487. [PMID: 28923931 DOI: 10.1073/pnas.1710625114] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Insulin receptors (IRs) on endothelial cells may have a role in the regulation of transport of circulating insulin to its target tissues; however, how this impacts on insulin action in vivo is unclear. Using mice with endothelial-specific inactivation of the IR gene (EndoIRKO), we find that in response to systemic insulin stimulation, loss of endothelial IRs caused delayed onset of insulin signaling in skeletal muscle, brown fat, hypothalamus, hippocampus, and prefrontal cortex but not in liver or olfactory bulb. At the level of the brain, the delay of insulin signaling was associated with decreased levels of hypothalamic proopiomelanocortin, leading to increased food intake and obesity accompanied with hyperinsulinemia and hyperleptinemia. The loss of endothelial IRs also resulted in a delay in the acute hypoglycemic effect of systemic insulin administration and impaired glucose tolerance. In high-fat diet-treated mice, knockout of the endothelial IRs accelerated development of systemic insulin resistance but not food intake and obesity. Thus, IRs on endothelial cells have an important role in transendothelial insulin delivery in vivo which differentially regulates the kinetics of insulin signaling and insulin action in peripheral target tissues and different brain regions. Loss of this function predisposes animals to systemic insulin resistance, overeating, and obesity.
Collapse
|
21
|
Gray SM, Aylor KW, Barrett EJ. Unravelling the regulation of insulin transport across the brain endothelial cell. Diabetologia 2017; 60:1512-1521. [PMID: 28601906 PMCID: PMC5534844 DOI: 10.1007/s00125-017-4285-4] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/23/2017] [Indexed: 11/28/2022]
Abstract
AIMS/HYPOTHESIS For circulating insulin to act on the brain it must cross the blood-brain barrier (BBB). Remarkably little is known about how circulating insulin crosses the BBB's highly restrictive brain endothelial cells (BECs). Therefore, we examined potential mechanisms regulating BEC insulin uptake, signalling and degradation during BEC transcytosis, and how transport is affected by a high-fat diet (HFD) and by astrocyte activity. METHODS 125I-TyrA14-insulin uptake and transcytosis, and the effects of insulin receptor (IR) blockade, inhibition of insulin signalling, astrocyte stimulation and an HFD were tested using purified isolated BECs (iBECs) in monoculture and co-cultured with astrocytes. RESULTS At physiological insulin concentrations, the IR, not the IGF-1 receptor, facilitated BEC insulin uptake, which required lipid raft-mediated endocytosis, but did not require insulin action on phosphoinositide-3-kinase (PI3K) or mitogen-activated protein kinase kinase (MEK). Feeding rats an HFD for 4 weeks decreased iBEC insulin uptake and increased NF-κB binding activity without affecting insulin PI3K signalling, IR expression or content, or insulin degrading enzyme expression. Using an in vitro BBB (co-culture of iBECs and astrocytes), we found insulin was not degraded during transcytosis, and that stimulating astrocytes with L-glutamate increased transcytosis, while inhibiting nitric oxide synthase decreased insulin transcytosis. CONCLUSIONS/INTERPRETATION Insulin crosses the BBB intact via an IR-specific, vesicle-mediated transport process in the BECs. HFD feeding, nitric oxide inhibition and astrocyte stimulation can regulate BEC insulin uptake and transcytosis.
Collapse
Affiliation(s)
- Sarah M Gray
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Kevin W Aylor
- Division of Endocrinology & Metabolism, Department of Medicine, University of Virginia, 450 Ray C. Hunt Drive, P.O. Box 801410, Charlottesville, VA, 22908, USA
| | - Eugene J Barrett
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA.
- Division of Endocrinology & Metabolism, Department of Medicine, University of Virginia, 450 Ray C. Hunt Drive, P.O. Box 801410, Charlottesville, VA, 22908, USA.
| |
Collapse
|
22
|
Solomon-Zemler R, Basel-Vanagaite L, Steier D, Yakar S, Mel E, Phillip M, Bazak L, Bercovich D, Werner H, de Vries L. A novel heterozygous IGF-1 receptor mutation associated with hypoglycemia. Endocr Connect 2017; 6. [PMID: 28649085 PMCID: PMC5551424 DOI: 10.1530/ec-17-0038] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Mutation in the insulin-like growth factor-1 receptor (IGF1R) gene is a rare cause for intrauterine and postnatal growth disorders. Patients identified with IGF1R mutations present with either normal or impaired glucose tolerance. None of the cases described so far showed hypoglycemia. We aimed to identify the genetic basis for small for gestational age, short stature and hypoglycemia over three generations in one family. The proband, a 9-year-old male, presented in infancy with recurrent hypoglycemic episodes, symmetric intrauterine growth retardation and postnatal growth retardation. Blood DNA samples from the patient, his parents, a maternal sister and maternal grandmother underwent Sanger sequencing of the IGF1R gene. Primary skin fibroblast cultures of the patient, his mother and age- and sex-matched control donors were used for gene expression and receptor functional analyses. We found a novel heterozygous mutation (c.94 + 1g > a, D1105E) affecting the splicing site of the IGF1R mRNA in the patient, his mother and his grandmother. Primary fibroblast cultures derived from the patient and his mother showed reduced proliferation and impaired activation of the IGF1R, evident by reduced IGF1R and AKT phosphorylation upon ligand binding. In conclusion, the newly identified heterozygous missense mutation in exon 1 of IGF1R (D1105E) results in impaired IGF1R function and is associated with small for gestational age, microcephaly and abnormal glucose metabolism. Further studies are required to understand the mechanisms by which this mutation leads to hypoglycemia.
Collapse
Affiliation(s)
- R Solomon-Zemler
- Sackler Faculty of MedicineTel Aviv University, Tel Aviv, Israel
| | - L Basel-Vanagaite
- Sackler Faculty of MedicineTel Aviv University, Tel Aviv, Israel
- Raphael Recanati Genetic InstituteRabin Medical Center - Beilinson Hospital, Petach Tikva, Israel
- Felsenstein Medical Research CenterPetach Tikva, Israel
- Pediatric GeneticsSchneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - D Steier
- Day Hospitalization DepartmentSchneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - S Yakar
- David B. Kriser Dental CenterDepartment of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York, USA
| | - E Mel
- Jesse Z. and Sara Lea Shafer Institute for Endocrinology and DiabetesSchneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - M Phillip
- Sackler Faculty of MedicineTel Aviv University, Tel Aviv, Israel
- Jesse Z. and Sara Lea Shafer Institute for Endocrinology and DiabetesSchneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - L Bazak
- Raphael Recanati Genetic InstituteRabin Medical Center - Beilinson Hospital, Petach Tikva, Israel
| | | | - H Werner
- Sackler Faculty of MedicineTel Aviv University, Tel Aviv, Israel
- Shalom and VardaYoran Institute for Human Genome ResearchTel Aviv University, Tel Aviv, Israel
| | - L de Vries
- Sackler Faculty of MedicineTel Aviv University, Tel Aviv, Israel
- Jesse Z. and Sara Lea Shafer Institute for Endocrinology and DiabetesSchneider Children's Medical Center of Israel, Petach Tikva, Israel
| |
Collapse
|
23
|
Saber H, Himali JJ, Beiser AS, Shoamanesh A, Pikula A, Roubenoff R, Romero JR, Kase CS, Vasan RS, Seshadri S. Serum Insulin-Like Growth Factor 1 and the Risk of Ischemic Stroke: The Framingham Study. Stroke 2017; 48:1760-1765. [PMID: 28596451 PMCID: PMC5505338 DOI: 10.1161/strokeaha.116.016563] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/04/2017] [Accepted: 05/04/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND PURPOSE Low insulin-like growth factor 1 (IGF-1) has been associated with increased risk of atherosclerosis and atrial fibrillation in cross-sectional studies. Yet, prospective data linking IGF-1 levels to the development of ischemic stroke remain inconclusive. We examined prospectively the association between serum IGF-1 levels and incident ischemic stroke. METHODS We measured serum IGF-1 levels in 757 elderly individuals (mean age 79±5, 62% women), free of prevalent stroke, from the Framingham original cohort participants at the 22nd examination cycle (1990-1994) and were followed up for the development of ischemic stroke. Cox models were used to relate IGF-1 levels to the risk for incident ischemic stroke, adjusted for potential confounders. RESULTS During a mean follow-up of 10.2 years, 99 individuals developed ischemic stroke. After adjustment for age, sex, and potential confounders, higher IGF-1 levels were associated with a lower risk of incident ischemic stroke, with subjects in the lowest quintile of IGF-1 levels having a 2.3-fold higher risk of incident ischemic stroke (95% confidence interval, 1.09-5.06; P=0.03) as compared with those in the top quintile. We observed an effect modification by diabetes mellitus and waist-hip ratio for the association between IGF-1 and ischemic stroke (P<0.1). In subgroup analyses, the effects were restricted to subjects with diabetics and those in top waist-hip ratio quartile, in whom each standard deviation increase in IGF-1 was associated with a 61% (hazard ratio, 0.39; 95% confidence interval, 0.20-0.78; P=0.007) and 41% (hazard ratio, 0.59; 95% confidence interval, 0.37-0.95; P=0.031) lower risk of incident ischemic stroke, respectively. CONCLUSIONS IGF-1 levels were inversely associated with ischemic stroke, especially among persons with insulin resistance.
Collapse
Affiliation(s)
- Hamidreza Saber
- From the Framingham Heart Study, MA (H.S., J.J.H., A.S.B., J.R.R., C.S.K., R.S.V., S.S.); Boston University School of Public Health, MA (J.J.H., A.S.B.); McMaster University/Population Health Research Institute, Hamilton, Ontario, Canada (A.S.); Department of Neurology, University of Toronto, Ontario, Canada (A.P.); Department of Medicine, Tufts University School of Medicine, Boston, MA (R.R.); Department of Neurology, Boston University School of Medicine, MA (J.J.H., A.S.B., J.R.R., C.S.K., R.S.V., S.S.); and Department of Neurology, Wayne State University School of Medicine, Detroit, MI (H.S.)
| | - Jayandra J Himali
- From the Framingham Heart Study, MA (H.S., J.J.H., A.S.B., J.R.R., C.S.K., R.S.V., S.S.); Boston University School of Public Health, MA (J.J.H., A.S.B.); McMaster University/Population Health Research Institute, Hamilton, Ontario, Canada (A.S.); Department of Neurology, University of Toronto, Ontario, Canada (A.P.); Department of Medicine, Tufts University School of Medicine, Boston, MA (R.R.); Department of Neurology, Boston University School of Medicine, MA (J.J.H., A.S.B., J.R.R., C.S.K., R.S.V., S.S.); and Department of Neurology, Wayne State University School of Medicine, Detroit, MI (H.S.)
| | - Alexa S Beiser
- From the Framingham Heart Study, MA (H.S., J.J.H., A.S.B., J.R.R., C.S.K., R.S.V., S.S.); Boston University School of Public Health, MA (J.J.H., A.S.B.); McMaster University/Population Health Research Institute, Hamilton, Ontario, Canada (A.S.); Department of Neurology, University of Toronto, Ontario, Canada (A.P.); Department of Medicine, Tufts University School of Medicine, Boston, MA (R.R.); Department of Neurology, Boston University School of Medicine, MA (J.J.H., A.S.B., J.R.R., C.S.K., R.S.V., S.S.); and Department of Neurology, Wayne State University School of Medicine, Detroit, MI (H.S.)
| | - Ashkan Shoamanesh
- From the Framingham Heart Study, MA (H.S., J.J.H., A.S.B., J.R.R., C.S.K., R.S.V., S.S.); Boston University School of Public Health, MA (J.J.H., A.S.B.); McMaster University/Population Health Research Institute, Hamilton, Ontario, Canada (A.S.); Department of Neurology, University of Toronto, Ontario, Canada (A.P.); Department of Medicine, Tufts University School of Medicine, Boston, MA (R.R.); Department of Neurology, Boston University School of Medicine, MA (J.J.H., A.S.B., J.R.R., C.S.K., R.S.V., S.S.); and Department of Neurology, Wayne State University School of Medicine, Detroit, MI (H.S.)
| | - Aleksandra Pikula
- From the Framingham Heart Study, MA (H.S., J.J.H., A.S.B., J.R.R., C.S.K., R.S.V., S.S.); Boston University School of Public Health, MA (J.J.H., A.S.B.); McMaster University/Population Health Research Institute, Hamilton, Ontario, Canada (A.S.); Department of Neurology, University of Toronto, Ontario, Canada (A.P.); Department of Medicine, Tufts University School of Medicine, Boston, MA (R.R.); Department of Neurology, Boston University School of Medicine, MA (J.J.H., A.S.B., J.R.R., C.S.K., R.S.V., S.S.); and Department of Neurology, Wayne State University School of Medicine, Detroit, MI (H.S.)
| | - Ronenn Roubenoff
- From the Framingham Heart Study, MA (H.S., J.J.H., A.S.B., J.R.R., C.S.K., R.S.V., S.S.); Boston University School of Public Health, MA (J.J.H., A.S.B.); McMaster University/Population Health Research Institute, Hamilton, Ontario, Canada (A.S.); Department of Neurology, University of Toronto, Ontario, Canada (A.P.); Department of Medicine, Tufts University School of Medicine, Boston, MA (R.R.); Department of Neurology, Boston University School of Medicine, MA (J.J.H., A.S.B., J.R.R., C.S.K., R.S.V., S.S.); and Department of Neurology, Wayne State University School of Medicine, Detroit, MI (H.S.)
| | - Jose R Romero
- From the Framingham Heart Study, MA (H.S., J.J.H., A.S.B., J.R.R., C.S.K., R.S.V., S.S.); Boston University School of Public Health, MA (J.J.H., A.S.B.); McMaster University/Population Health Research Institute, Hamilton, Ontario, Canada (A.S.); Department of Neurology, University of Toronto, Ontario, Canada (A.P.); Department of Medicine, Tufts University School of Medicine, Boston, MA (R.R.); Department of Neurology, Boston University School of Medicine, MA (J.J.H., A.S.B., J.R.R., C.S.K., R.S.V., S.S.); and Department of Neurology, Wayne State University School of Medicine, Detroit, MI (H.S.)
| | - Carlos S Kase
- From the Framingham Heart Study, MA (H.S., J.J.H., A.S.B., J.R.R., C.S.K., R.S.V., S.S.); Boston University School of Public Health, MA (J.J.H., A.S.B.); McMaster University/Population Health Research Institute, Hamilton, Ontario, Canada (A.S.); Department of Neurology, University of Toronto, Ontario, Canada (A.P.); Department of Medicine, Tufts University School of Medicine, Boston, MA (R.R.); Department of Neurology, Boston University School of Medicine, MA (J.J.H., A.S.B., J.R.R., C.S.K., R.S.V., S.S.); and Department of Neurology, Wayne State University School of Medicine, Detroit, MI (H.S.)
| | - Ramachandran S Vasan
- From the Framingham Heart Study, MA (H.S., J.J.H., A.S.B., J.R.R., C.S.K., R.S.V., S.S.); Boston University School of Public Health, MA (J.J.H., A.S.B.); McMaster University/Population Health Research Institute, Hamilton, Ontario, Canada (A.S.); Department of Neurology, University of Toronto, Ontario, Canada (A.P.); Department of Medicine, Tufts University School of Medicine, Boston, MA (R.R.); Department of Neurology, Boston University School of Medicine, MA (J.J.H., A.S.B., J.R.R., C.S.K., R.S.V., S.S.); and Department of Neurology, Wayne State University School of Medicine, Detroit, MI (H.S.)
| | - Sudha Seshadri
- From the Framingham Heart Study, MA (H.S., J.J.H., A.S.B., J.R.R., C.S.K., R.S.V., S.S.); Boston University School of Public Health, MA (J.J.H., A.S.B.); McMaster University/Population Health Research Institute, Hamilton, Ontario, Canada (A.S.); Department of Neurology, University of Toronto, Ontario, Canada (A.P.); Department of Medicine, Tufts University School of Medicine, Boston, MA (R.R.); Department of Neurology, Boston University School of Medicine, MA (J.J.H., A.S.B., J.R.R., C.S.K., R.S.V., S.S.); and Department of Neurology, Wayne State University School of Medicine, Detroit, MI (H.S.).
| |
Collapse
|
24
|
Lee WL, Klip A. Endothelial Transcytosis of Insulin: Does It Contribute to Insulin Resistance? Physiology (Bethesda) 2017; 31:336-45. [PMID: 27511460 DOI: 10.1152/physiol.00010.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Most research on insulin resistance has focused on impaired signaling at the level of target tissues like skeletal muscle. Insulin delivery is also important and includes recruitment and perfusion of capillaries bearing insulin, but also the transit of insulin across the capillary endothelium. The mechanisms of this second stage (insulin transcytosis) and whether it contributes to insulin resistance remain uncertain.
Collapse
Affiliation(s)
- Warren L Lee
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada; Department of Medicine, University of Toronto, Toronto, Canada; Department of Biochemistry, University of Toronto, Toronto, Canada; Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; and
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada; Department of Biochemistry, University of Toronto, Toronto, Canada; Paediatrics, and Physiology, University of Toronto, Toronto, Canada
| |
Collapse
|
25
|
Fu Z, Wu J, Nesil T, Li MD, Aylor KW, Liu Z. Long-term high-fat diet induces hippocampal microvascular insulin resistance and cognitive dysfunction. Am J Physiol Endocrinol Metab 2017; 312:E89-E97. [PMID: 27899343 PMCID: PMC5336564 DOI: 10.1152/ajpendo.00297.2016] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 11/04/2016] [Accepted: 11/21/2016] [Indexed: 12/20/2022]
Abstract
Insulin action on hippocampus improves cognitive function, and obesity and type 2 diabetes are associated with decreased cognitive function. Cerebral microvasculature plays a critical role in maintaining cerebral vitality and function by supplying nutrients, oxygen, and hormones such as insulin to cerebral parenchyma, including hippocampus. In skeletal muscle, insulin actively regulates microvascular opening and closure, and this action is impaired in the insulin-resistant states. To examine insulin's action on hippocampal microvasculature and parenchyma and the impact of diet-induced obesity, we determined cognitive function and microvascular insulin responses, parenchyma insulin responses, and capillary density in the hippocampus in 2- and 8-mo-old rats on chow diet and 8-mo-old rats on a long-term high-fat diet (6 mo). Insulin infusion increased hippocampal microvascular perfusion in rats on chow diet by ~80-90%. High-fat diet feeding completely abolished insulin-mediated microvascular responses and protein kinase B phosphorylation but did not alter the capillary density in the hippocampus. This was associated with a significantly decreased cognitive function assessed using both the two-trial spontaneous alternation behavior test and the novel object recognition test. As the microvasculature provides the needed endothelial surface area for delivery of nutrients, oxygen, and insulin to hippocampal parenchyma, we conclude that hippocampal microvascular insulin resistance may play a critical role in the development of cognitive impairment seen in obesity and diabetes. Our results suggest that improvement in hippocampal microvascular insulin sensitivity might help improve or reverse cognitive function in the insulin-resistant states.
Collapse
Affiliation(s)
- Zhuo Fu
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia
| | - Jing Wu
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia
- Department of Endocrinology, Central South University Xiangya Hospital, Changsha, Hunan, China; and
| | - Tanseli Nesil
- Department of Psychiatry, University of Virginia Health System, Charlottesville, Virginia
| | - Ming D Li
- Department of Psychiatry, University of Virginia Health System, Charlottesville, Virginia
| | - Kevin W Aylor
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia
| | - Zhenqi Liu
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia;
| |
Collapse
|
26
|
Gralle M. The neuronal insulin receptor in its environment. J Neurochem 2016; 140:359-367. [PMID: 27889917 DOI: 10.1111/jnc.13909] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 10/31/2016] [Accepted: 11/21/2016] [Indexed: 01/01/2023]
Abstract
Insulin is known mainly for its effects in peripheral tissues, such as the liver, skeletal muscles and adipose tissue, where the activation of the insulin receptor (IR) has both short-term and long-term effects. Insulin and the IR are also present in the brain, and since there is evidence that neuronal insulin signaling regulates synaptic plasticity and that it is impaired in disease, this pathway might be the key to protection or reversal of symptoms, especially in Alzheimer's disease. However, there are controversies about the importance of the neuronal IR, partly because biophysical data on its activation and signaling are much less complete than for the peripheral IR. This review briefly summarizes the neuronal IR signaling in health and disease, and then focuses on known differences between the neuronal and peripheral IR with regard to alternative splicing and glycosylation, and lack of data with respect to phosphorylation and membrane subdomain localization. Particularities in the neuronal IR itself and its environment may have consequences for downstream signaling and impact synaptic plasticity. Furthermore, establishing the relative importance of insulin signaling through IR or through hybrids with its homolog, the insulin-like growth factor 1 receptor, is crucial for evaluating the consequences of brain IR activation. An improved biophysical understanding of the neuronal IR may help predict the consequences of insulin-targeted interventions.
Collapse
Affiliation(s)
- Matthias Gralle
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
27
|
van Beijnum JR, Pieters W, Nowak-Sliwinska P, Griffioen AW. Insulin-like growth factor axis targeting in cancer and tumour angiogenesis - the missing link. Biol Rev Camb Philos Soc 2016; 92:1755-1768. [PMID: 27779364 DOI: 10.1111/brv.12306] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 09/15/2016] [Accepted: 09/21/2016] [Indexed: 12/14/2022]
Abstract
Numerous molecular players in the process of tumour angiogenesis have been shown to offer potential for therapeutic targeting. Initially denoted to be involved in malignant transformation and tumour progression, the insulin-like growth factor (IGF) signalling axis has been subject to therapeutic interference, albeit with limited clinical success. More recently, IGFs and their receptors have received attention for their contribution to tumour angiogenesis, which offers novel therapeutic opportunities. Here we review the contribution of this signalling axis to tumour angiogenesis, the mechanisms of resistance to therapy and the interplay with other pro-angiogenic pathways, to offer insight in the renewed interest in the application of IGF axis targeting agents in anti-cancer combination therapies.
Collapse
Affiliation(s)
- Judy R van Beijnum
- Department of Medical Oncology, Angiogenesis Laboratory, VU University Medical Center, PO box 7057, 1007 MB, Amsterdam, The Netherlands
| | - Wietske Pieters
- Department of Medical Oncology, Angiogenesis Laboratory, VU University Medical Center, PO box 7057, 1007 MB, Amsterdam, The Netherlands
| | - Patrycja Nowak-Sliwinska
- School of Pharmaceutical Sciences, University of Geneva (UNIGE), Rue Michel-Servet 1, 1211 Geneva 4, Switzerland
| | - Arjan W Griffioen
- Department of Medical Oncology, Angiogenesis Laboratory, VU University Medical Center, PO box 7057, 1007 MB, Amsterdam, The Netherlands
| |
Collapse
|
28
|
Najem D, Bamji-Mirza M, Yang Z, Zhang W. Aβ-Induced Insulin Resistance and the Effects of Insulin on the Cholesterol Synthesis Pathway and Aβ Secretion in Neural Cells. Neurosci Bull 2016; 32:227-38. [PMID: 27207326 DOI: 10.1007/s12264-016-0034-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 04/21/2016] [Indexed: 10/21/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by amyloid-β (Aβ) toxicity, tau pathology, insulin resistance, neuroinflammation, and dysregulation of cholesterol homeostasis, all of which play roles in neurodegeneration. Insulin has polytrophic effects on neurons and may be at the center of these pathophysiological changes. In this study, we investigated possible relationships among insulin signaling and cholesterol biosynthesis, along with the effects of Aβ42 on these pathways in vitro. We found that neuroblastoma 2a (N2a) cells transfected with the human gene encoding amyloid-β protein precursor (AβPP) (N2a-AβPP) produced Aβ and exhibited insulin resistance by reduced p-Akt and a suppressed cholesterol-synthesis pathway following insulin treatment, and by increased phosphorylation of insulin receptor subunit-1 at serine 612 (p-IRS-S612) as compared to parental N2a cells. Treatment of human neuroblastoma SH-SY5Y cells with Aβ42 also increased p-IRS-S612, suggesting that Aβ42 is responsible for insulin resistance. The insulin resistance was alleviated when N2a-AβPP cells were treated with higher insulin concentrations. Insulin increased Aβ release from N2a-AβPP cells, by which it may promote Aβ clearance. Insulin increased cholesterol-synthesis gene expression in SH-SY5Y and N2a cells, including 24-dehydrocholesterol reductase (DHCR24) and 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGCR) through sterol-regulatory element-binding protein-2 (SREBP2). While Aβ42-treated SH-SY5Y cells exhibited increased HMGCR expression and c-Jun phosphorylation as pro-inflammatory responses, they also showed down-regulation of neuro-protective/anti-inflammatory DHCR24. These results suggest that Aβ42 may cause insulin resistance, activate JNK for c-Jun phosphorylation, and lead to dysregulation of cholesterol homeostasis, and that enhancing insulin signaling may relieve the insulin-resistant phenotype and the dysregulated cholesterol-synthesis pathway to promote Aβ release for clearance from neural cells.
Collapse
Affiliation(s)
- Dema Najem
- Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H8M5, Canada.,Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, K1A0R6, Canada
| | - Michelle Bamji-Mirza
- Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H8M5, Canada.,Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, K1A0R6, Canada
| | - Ze Yang
- Beijing Hospital, Institute of Geriatrics-Chinese Health Ministry, Beijing, 100730, China
| | - Wandong Zhang
- Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H8M5, Canada. .,Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, K1A0R6, Canada.
| |
Collapse
|
29
|
Wang H, Wang AX, Aylor K, Barrett EJ. Caveolin-1 phosphorylation regulates vascular endothelial insulin uptake and is impaired by insulin resistance in rats. Diabetologia 2015; 58:1344-53. [PMID: 25748795 PMCID: PMC4417063 DOI: 10.1007/s00125-015-3546-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 02/13/2015] [Indexed: 10/25/2022]
Abstract
AIMS/HYPOTHESIS As insulin entry into muscle interstitium is rate-limiting for its overall peripheral action, defining the route and regulation of its entry is critical. Caveolin-1 is required for caveola formation in vascular endothelial cells (ECs) and for EC insulin uptake. Whether this requirement reflects simply the need for caveola availability or involves a more active role for caveolae/caveolin-1 is not known. Here, we examined the role of insulin-stimulated tyrosine 14 (Tyr(14))-caveolin-1 phosphorylation in mediating EC insulin uptake and the role of cellular Src-kinase (cSrc), TNF-α/IL-6 and high fat diet (HFD) in regulating this process. METHODS Freshly isolated ECs from normal or HFD-fed rats and/or cultured ECs were treated with FITC-labelled or regular insulin with or without a Src or phosphotidylinositol-3-kinase inhibitor, TNF-α or IL-6, or transfecting FLAG-tagged wild-type (WT) or mutant (Y14F) caveolin-1. Tyr(14)-caveolin-1/Tyr(416) cSrc phosphorylation and FITC-insulin uptake were quantified by immunostaining and/or western blots. RESULTS Insulin stimulated Tyr(14)-caveolin-1 phosphorylation during EC insulin uptake. Inhibiting cSrc, but not phosphotidylinositol-3-kinase, reduced insulin-stimulated caveolin-1 phosphorylation. Furthermore, inhibiting cSrc reduced FITC-insulin uptake by ∼50%. Overexpression of caveolin-1Y14F inhibited, while overexpression of WT caveolin-1 increased, FITC-insulin uptake. Exposure of ECs to TNF-α or IL-6, or to 1-week HFD feeding eliminated insulin-stimulated caveolin-1 phosphorylation and inhibited FITC-insulin uptake to a similar extent. CONCLUSIONS/INTERPRETATION Insulin stimulation of its own uptake requires caveolin-1 phosphorylation and Src-kinase activity. HFD in vivo and proinflammatory cytokines in vitro both inhibit this process.
Collapse
Affiliation(s)
- Hong Wang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Virginia Health System, 450 Ray C. Hunt Drive, Box 801410, Charlottesville, VA, 22908, USA,
| | | | | | | |
Collapse
|
30
|
Zheng C, Liu Z. Vascular function, insulin action, and exercise: an intricate interplay. Trends Endocrinol Metab 2015; 26:297-304. [PMID: 25735473 PMCID: PMC4450131 DOI: 10.1016/j.tem.2015.02.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 01/29/2015] [Accepted: 02/03/2015] [Indexed: 01/04/2023]
Abstract
Insulin enhances the compliance of conduit arteries, relaxes resistance arterioles to increase tissue blood flow, and dilates precapillary arterioles to expand muscle microvascular blood volume. These actions are impaired in the insulin resistant states. Exercise ameliorates endothelial dysfunction and improves insulin responses in insulin resistant patients, but the precise underlying mechanisms remain unclear. The microvasculature critically regulates insulin action in muscle by modulating insulin delivery to the capillaries nurturing the myocytes and trans-endothelial insulin transport. Recent data suggest that exercise may exert its insulin-sensitizing effect via recruiting muscle microvasculature to increase insulin delivery to and action in muscle. The current review focuses on how the interplay among exercise, insulin action, and the vasculature contributes to exercise-mediated insulin sensitization in muscle.
Collapse
Affiliation(s)
- Chao Zheng
- Diabetes Center and Department of Endocrinology, the Second Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Zhenqi Liu
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA, USA.
| |
Collapse
|
31
|
Abstract
Endothelial cells line blood vessels and modulate vascular tone, thrombosis, inflammatory responses and new vessel formation. They are implicated in many disease processes including atherosclerosis and cancer. IGFs play a significant role in the physiology of endothelial cells by promoting migration, tube formation and production of the vasodilator nitric oxide. These actions are mediated by the IGF1 and IGF2/mannose 6-phosphate receptors and are modulated by a family of high-affinity IGF binding proteins. IGFs also increase the number and function of endothelial progenitor cells, which may contribute to protection from atherosclerosis. IGFs promote angiogenesis, and dysregulation of the IGF system may contribute to this process in cancer and eye diseases including retinopathy of prematurity and diabetic retinopathy. In some situations, IGF deficiency appears to contribute to endothelial dysfunction, whereas IGF may be deleterious in others. These differences may be due to tissue-specific endothelial cell phenotypes or IGFs having distinct roles in different phases of vascular disease. Further studies are therefore required to delineate the therapeutic potential of IGF system modulation in pathogenic processes.
Collapse
Affiliation(s)
- Leon A Bach
- Department of Medicine (Alfred)Monash University, Prahran 3181, AustraliaDepartment of Endocrinology and DiabetesAlfred Hospital, Commercial Road, Melbourne 3004, Australia Department of Medicine (Alfred)Monash University, Prahran 3181, AustraliaDepartment of Endocrinology and DiabetesAlfred Hospital, Commercial Road, Melbourne 3004, Australia
| |
Collapse
|
32
|
Westermeier F, Salomón C, Farías M, Arroyo P, Fuenzalida B, Sáez T, Salsoso R, Sanhueza C, Guzmán‐Gutiérrez E, Pardo F, Leiva A, Sobrevia L. Insulin requires normal expression and signaling of insulin receptor A to reverse gestational diabetes‐reduced adenosine transport in human umbilical vein endothelium. FASEB J 2015; 29:37-49. [DOI: 10.1096/fj.14-254219] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Francisco Westermeier
- Cellular and Molecular Physiology Laboratory (CMPL)Division of Obstetrics and GynaecologySchool of MedicineFaculty of MedicinePontificia Universidad Católica de ChileSantiagoChile
- Advanced Center for Chronic Diseases (ACCDIS)Faculty of Chemical & Pharmaceutical SciencesUniversidad de ChileSantiagoChile
| | - Carlos Salomón
- Cellular and Molecular Physiology Laboratory (CMPL)Division of Obstetrics and GynaecologySchool of MedicineFaculty of MedicinePontificia Universidad Católica de ChileSantiagoChile
- University of Queensland Centre for Clinical Research (UQCCR)Faculty of Medicine and Biomedical SciencesUniversity of QueenslandHerstonQueenslandAustralia
| | - Marcelo Farías
- Cellular and Molecular Physiology Laboratory (CMPL)Division of Obstetrics and GynaecologySchool of MedicineFaculty of MedicinePontificia Universidad Católica de ChileSantiagoChile
| | - Pablo Arroyo
- Cellular and Molecular Physiology Laboratory (CMPL)Division of Obstetrics and GynaecologySchool of MedicineFaculty of MedicinePontificia Universidad Católica de ChileSantiagoChile
| | - Bárbara Fuenzalida
- Cellular and Molecular Physiology Laboratory (CMPL)Division of Obstetrics and GynaecologySchool of MedicineFaculty of MedicinePontificia Universidad Católica de ChileSantiagoChile
| | - Tamara Sáez
- Cellular and Molecular Physiology Laboratory (CMPL)Division of Obstetrics and GynaecologySchool of MedicineFaculty of MedicinePontificia Universidad Católica de ChileSantiagoChile
| | - Rocío Salsoso
- Cellular and Molecular Physiology Laboratory (CMPL)Division of Obstetrics and GynaecologySchool of MedicineFaculty of MedicinePontificia Universidad Católica de ChileSantiagoChile
| | - Carlos Sanhueza
- Cellular and Molecular Physiology Laboratory (CMPL)Division of Obstetrics and GynaecologySchool of MedicineFaculty of MedicinePontificia Universidad Católica de ChileSantiagoChile
| | - Enrique Guzmán‐Gutiérrez
- Cellular and Molecular Physiology Laboratory (CMPL)Division of Obstetrics and GynaecologySchool of MedicineFaculty of MedicinePontificia Universidad Católica de ChileSantiagoChile
- Faculty of Health SciencesUniversidad San SebastiánConcepciónChile
| | - Fabián Pardo
- Cellular and Molecular Physiology Laboratory (CMPL)Division of Obstetrics and GynaecologySchool of MedicineFaculty of MedicinePontificia Universidad Católica de ChileSantiagoChile
| | - Andrea Leiva
- Cellular and Molecular Physiology Laboratory (CMPL)Division of Obstetrics and GynaecologySchool of MedicineFaculty of MedicinePontificia Universidad Católica de ChileSantiagoChile
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL)Division of Obstetrics and GynaecologySchool of MedicineFaculty of MedicinePontificia Universidad Católica de ChileSantiagoChile
- University of Queensland Centre for Clinical Research (UQCCR)Faculty of Medicine and Biomedical SciencesUniversity of QueenslandHerstonQueenslandAustralia
- Faculty of PharmacyUniversidad de SevillaSevilleSpain
| |
Collapse
|
33
|
Pandey G, Makhija E, George N, Chakravarti B, Godbole MM, Ecelbarger CM, Tiwari S. Insulin regulates nitric oxide production in the kidney collecting duct cells. J Biol Chem 2014; 290:5582-91. [PMID: 25533472 DOI: 10.1074/jbc.m114.592741] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The kidney is an important organ for arterial blood pressure (BP) maintenance. Reduced NO generation in the kidney is associated with hypertension in insulin resistance. NO is a critical regulator of vascular tone; however, whether insulin regulates NO production in the renal inner medullary collecting duct (IMCD), the segment with the greatest enzymatic activity for NO production in kidney, is not clear. Using an NO-sensitive 4-amino-5-methylamino-2',7'-difluorofluorescein (DAF-FM) fluorescent dye, we found that insulin increased NO production in mouse IMCD cells (mIMCD) in a time- and dose-dependent manner. A concomitant dose-dependent increase in the NO metabolite (NOx) was also observed in the medium from insulin-stimulated cells. NO production peaked in mIMCD cells at a dose of 100 nm insulin with simultaneously increased NOx levels in the medium. At this dose, insulin significantly increased p-eNOS(Ser1177) levels in mIMCD cells. Pretreatment of cells with a PI 3-kinase inhibitor or insulin receptor silencing with RNA interference abolished these effects of insulin, whereas insulin-like growth factor-1 receptor (IGF-1R) silencing had no effect. We also showed that chronic insulin infusion to normal C57BL/6J mice resulted in increased endothelial NOS (eNOS) protein levels and NO production in the inner medulla. However, insulin-infused IRKO mice, with targeted deletion of insulin receptor from tubule epithelial cells of the kidney, had ∼50% reduced eNOS protein levels in their inner medulla along with a significant rise in BP relative to WT littermates. We have previously reported increased baseline BP and reduced urine NOx in IRKO mice. Thus, reduced insulin receptor signaling in IMCD could contribute to hypertension in the insulin-resistant state.
Collapse
Affiliation(s)
- Gaurav Pandey
- From the Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India and
| | - Ekta Makhija
- From the Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India and
| | - Nelson George
- From the Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India and
| | - Bandana Chakravarti
- From the Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India and
| | - Madan M Godbole
- From the Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India and
| | - Carolyn M Ecelbarger
- the Division of Endocrinology and Metabolism, Department of Medicine, Georgetown University, Washington, D. C. 2007
| | - Swasti Tiwari
- From the Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India and
| |
Collapse
|
34
|
Higashi Y, Quevedo HC, Tiwari S, Sukhanov S, Shai SY, Anwar A, Delafontaine P. Interaction between insulin-like growth factor-1 and atherosclerosis and vascular aging. FRONTIERS OF HORMONE RESEARCH 2014; 43:107-24. [PMID: 24943302 DOI: 10.1159/000360571] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The process of vascular aging encompasses alterations in the function of endothelial (ECs) and vascular smooth muscle cells (VSMCs) via oxidation, inflammation, cell senescence and epigenetic modifications, increasing the probability of atherosclerosis. Aged vessels exhibit decreased endothelial antithrombogenic properties, increased reactive oxygen species generation, inflammatory signaling and migration of VSMCs to the subintimal space, impaired angiogenesis and increased elastin degradation. The key initiating step in atherogenesis is subendothelial accumulation of apolipoprotein B-containing low-density lipoproteins resulting in activation of ECs and recruitment of monocytes. Activated ECs secrete 'chemokines' that interact with cognate chemokine receptors on monocytes and promote directional migration. Recruitment of immune cells establishes a proinflammatory status, further causing elevated oxidative stress, which in turn triggers a series of events including apoptotic or necrotic death of vascular and nonvascular cells. Increased oxidative stress is also considered to be a key factor in mechanisms of aging-associated changes in tissue integrity and function. Experimental evidence indicates that insulin-like growth factor-1 exerts antioxidant, anti-inflammatory and pro-survival effects on the vasculature, reducing atherosclerotic plaque burden and promoting features of atherosclerotic plaque stability.
Collapse
Affiliation(s)
- Yusuke Higashi
- Tulane University School of Medicine, Tulane University Heart and Vascular Institute, New Orleans, La., USA
| | | | | | | | | | | | | |
Collapse
|
35
|
|
36
|
Fu Z, Zhao L, Aylor KW, Carey RM, Barrett EJ, Liu Z. Angiotensin-(1-7) recruits muscle microvasculature and enhances insulin's metabolic action via mas receptor. Hypertension 2014; 63:1219-27. [PMID: 24711523 DOI: 10.1161/hypertensionaha.113.03025] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Angiotensin-(1-7) [Ang-(1-7)], an endogenous ligand for the G protein-coupled receptor Mas, exerts both vasodilatory and insulin-sensitizing effects. In skeletal muscle, relaxation of precapillary arterioles recruits microvasculature and increases the endothelial surface area available for nutrient and hormone exchanges. To assess whether Ang-(1-7) recruits microvasculature and enhances insulin action in muscle, overnight-fasted adult rats received an intravenous infusion of Ang-(1-7) (0, 10, or 100 ng/kg per minute) for 150 minutes with or without a simultaneous infusion of the Mas inhibitor A-779 and a superimposition of a euglycemic insulin clamp (3 mU/kg per minute) from 30 to 150 minutes. Hind limb muscle microvascular blood volume, microvascular flow velocity, and microvascular blood flow were determined. Myographic changes in tension were measured on preconstricted distal saphenous artery. Ang-(1-7) dose-dependently relaxed the saphenous artery (P<0.05) ex vivo. This effect was potentiated by insulin (P<0.01) and abolished by either endothelium denudement or Mas inhibition. Systemic infusion of Ang-(1-7) rapidly increased muscle microvascular blood volume and microvascular blood flow (P<0.05, each) without altering microvascular flow velocity. Insulin infusion alone increased muscle microvascular blood volume by 60% to 70% (P<0.05). Adding insulin to the Ang-(1-7) infusion further increased muscle microvascular blood volume and microvascular blood flow (≈2.5 fold; P<0.01). These were associated with a significant increase in insulin-mediated glucose disposal and muscle protein kinase B and extracellular signal-regulated kinase 1/2 phosphorylation. A-779 pretreatment blunted the microvascular and insulin-sensitizing effects of Ang-(1-7). We conclude that Ang-(1-7) by activating Mas recruits muscle microvasculature and enhances the metabolic action of insulin. These effects may contribute to the cardiovascular protective responses associated with Mas activation and explain the insulin-sensitizing action of Ang-(1-7).
Collapse
Affiliation(s)
- Zhuo Fu
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, PO Box 801410, Charlottesville, VA 22908.
| | | | | | | | | | | |
Collapse
|
37
|
Othman EM, Hintzsche H, Stopper H. Signaling steps in the induction of genomic damage by insulin in colon and kidney cells. Free Radic Biol Med 2014; 68:247-57. [PMID: 24355212 DOI: 10.1016/j.freeradbiomed.2013.12.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 11/20/2013] [Accepted: 12/09/2013] [Indexed: 12/22/2022]
Abstract
Diabetes mellitus (DM), a disease with almost 350 million people affected worldwide, will be the seventh leading cause of death by 2030. Diabetic patients develop various types of complications, among them an increased rate of malignancies. Studies reported the strong correlation between DM and several cancer types, of which colon and kidney cancers are the most common. Hyperinsulinemia, the high insulin blood level characteristic of early diabetes type 2, was identified as a risk factor for cancer development. In previous studies, we showed that an elevated insulin level can induce oxidative stress, resulting in DNA damage in colon cells in vitro and in kidney cells in vitro and in vivo. In the present study, we elucidate the signaling pathway of insulin-mediated genotoxicity, which is effective through oxidative stress induction in colon and kidney. The signaling mechanism is starting by phosphorylation of the insulin and insulin-like growth factor-1 receptors, followed by activation of phosphatidylinositide 3-kinase (PI3K), which in turn activates AKT. Subsequently, mitochondria and nicotinamide adenine dinucleotide phosphate oxidase (NADPH) isoforms (Nox1 and Nox4 in colon and kidney, respectively) are activated for reactive oxygen species (ROS) production, and the resulting excess ROS can attack the DNA, causing DNA oxidation. We conclude that hyperinsulinemia represents an important risk factor for cancer initiation or progression as well as a target for cancer prevention in diabetic patients.
Collapse
Affiliation(s)
- Eman Maher Othman
- Institute of Pharmacology and Toxicology, University of Wuerzburg, D-97078 Wuerzburg, Germany; Department of Analytical Chemistry, Faculty of Pharmacy, University of El-Minia, 61519 El-Minia, Egypt
| | - Henning Hintzsche
- Institute of Pharmacology and Toxicology, University of Wuerzburg, D-97078 Wuerzburg, Germany
| | - Helga Stopper
- Institute of Pharmacology and Toxicology, University of Wuerzburg, D-97078 Wuerzburg, Germany.
| |
Collapse
|
38
|
Genders AJ, Frison V, Abramson SR, Barrett EJ. Endothelial cells actively concentrate insulin during its transendothelial transport. Microcirculation 2014; 20:434-9. [PMID: 23350546 DOI: 10.1111/micc.12044] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 01/23/2013] [Indexed: 01/07/2023]
Abstract
OBJECTIVE We examined insulins uptake and transendothelial transport by endothelial cells in order to: (i) ascertain whether insulin accumulates within the cells to concentrations greater than in the media; (ii) compare trans endothelial insulin transport to that of inulin (using the latter as a tracer for passive transport or leaked); and; (iii) determine whether insulins transported depended on insulin action. METHODS Using 125I-insulin at physiologic concentrations we measured both the uptake and trans endothelial transport of insulin by bovine aortic endothelial cells and measured cell volume using tritiated 3-O-methylglucose. RESULTS Bovine aortic endothelial cells accumulate insulin to > five-fold above the media concentrations and the trans endothelial transport of insulin, but not inulin, is saturable and requires intact PI-3-kinase and MEK signaling. CONCLUSION The insulin receptor and downstream signaling from the receptor regulates endothelial insulin transport. Insulin is accumulated against a concentration gradient by the endothelial cell. We suggest that insulin uptake is rate limiting for insulin trans endothelial transport.
Collapse
Affiliation(s)
- Amanda J Genders
- Division of Endocrinology, Department of Medicine, University of Virginia, School of Medicine, Charlottesville, VA 22908, USA
| | | | | | | |
Collapse
|
39
|
Ellis BC, Graham LD, Molloy PL. CRNDE, a long non-coding RNA responsive to insulin/IGF signaling, regulates genes involved in central metabolism. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1843:372-86. [PMID: 24184209 DOI: 10.1016/j.bbamcr.2013.10.016] [Citation(s) in RCA: 170] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 10/04/2013] [Accepted: 10/21/2013] [Indexed: 12/18/2022]
Abstract
Colorectal neoplasia differentially expressed (CRNDE) is a novel gene that is activated early in colorectal cancer but whose regulation and functions are unknown. CRNDE transcripts are recognized as long non-coding RNAs (lncRNAs), which potentially interact with chromatin-modifying complexes to regulate gene expression via epigenetic changes. Complex alternative splicing results in numerous transcripts from this gene, and we have identified novel transcripts containing a highly-conserved sequence within intron 4 ("gVC-In4"). In colorectal cancer cells, we demonstrate that treatment with insulin and insulin-like growth factors (IGF) repressed CRNDE nuclear transcripts, including those encompassing gVC-In4. These repressive effects were negated by use of inhibitors against either the PI3K/Akt/mTOR pathway or Raf/MAPK pathway, suggesting CRNDE is a downstream target of both signaling cascades. Expression array analyses revealed that siRNA-mediated knockdown of gVC-In4 transcripts affected the expression of many genes, which showed correlation with insulin/IGF signaling pathway components and responses, including glucose and lipid metabolism. Some of the genes are identical to those affected by insulin treatment in the same cell line. The results suggest that CRNDE expression promotes the metabolic changes by which cancer cells switch to aerobic glycolysis (Warburg effect). This is the first report of a lncRNA regulated by insulin/IGFs, and our findings indicate a role for CRNDE nuclear transcripts in regulating cellular metabolism which may correlate with their upregulation in colorectal cancer.
Collapse
Affiliation(s)
- Blake C Ellis
- CSIRO Animal, Food and Health Sciences, Preventative Health Flagship, Commonwealth Scientific and Industrial Research Organization, Sydney, NSW 2113 Australia.
| | - Lloyd D Graham
- CSIRO Animal, Food and Health Sciences, Preventative Health Flagship, Commonwealth Scientific and Industrial Research Organization, Sydney, NSW 2113 Australia.
| | - Peter L Molloy
- CSIRO Animal, Food and Health Sciences, Preventative Health Flagship, Commonwealth Scientific and Industrial Research Organization, Sydney, NSW 2113 Australia.
| |
Collapse
|
40
|
Cubbon RM, Mercer BN, Sengupta A, Kearney MT. Importance of insulin resistance to vascular repair and regeneration. Free Radic Biol Med 2013; 60:246-63. [PMID: 23466555 DOI: 10.1016/j.freeradbiomed.2013.02.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Revised: 02/22/2013] [Accepted: 02/23/2013] [Indexed: 01/14/2023]
Abstract
Metabolic insulin resistance is apparent across a spectrum of clinical disorders, including obesity and diabetes, and is characterized by an adverse clustering of cardiovascular risk factors related to abnormal cellular responses to insulin. These disorders are becoming increasingly prevalent and represent a major global public health concern because of their association with significant increases in atherosclerosis-related mortality. Endogenous repair mechanisms are thought to retard the development of vascular disease, and a growing evidence base supports the adverse impact of the insulin-resistant phenotype upon indices of vascular repair. Beyond the impact of systemic metabolic changes, emerging data from murine studies also provide support for abnormal insulin signaling at the level of vascular cells in retarding vascular repair. Interrelated pathophysiological factors, including reduced nitric oxide bioavailability, oxidative stress, altered growth factor activity, and abnormal intracellular signaling, are likely to act in conjunction to impede vascular repair while also driving vascular damage. Understanding of these processes is shaping novel therapeutic paradigms that aim to promote vascular repair and regeneration, either by recruiting endogenous mechanisms or by the administration of cell-based therapies.
Collapse
Affiliation(s)
- Richard M Cubbon
- Multidisciplinary Cardiovascular Research Centre, LIGHT Laboratories, The University of Leeds, Leeds LS2 9JT, UK.
| | | | | | | |
Collapse
|
41
|
Fu Z, Zhao L, Chai W, Dong Z, Cao W, Liu Z. Ranolazine recruits muscle microvasculature and enhances insulin action in rats. J Physiol 2013; 591:5235-49. [PMID: 23798495 DOI: 10.1113/jphysiol.2013.257246] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Ranolazine, an anti-anginal compound, has been shown to significantly improve glycaemic control in large-scale clinical trials, and short-term ranolazine treatment is associated with an improvement in myocardial blood flow. As microvascular perfusion plays critical roles in insulin delivery and action, we aimed to determine if ranolazine could improve muscle microvascular blood flow, thereby increasing muscle insulin delivery and glucose use. Overnight-fasted, anaesthetized Sprague-Dawley rats were used to determine the effects of ranolazine on microvascular recruitment using contrast-enhanced ultrasound, insulin action with euglycaemic hyperinsulinaemic clamp, and muscle insulin uptake using (125)I-insulin. Ranolazine's effects on endothelial nitric oxide synthase (eNOS) phosphorylation, cAMP generation and endothelial insulin uptake were determined in cultured endothelial cells. Ranolazine-induced myographical changes in tension were determined in isolated distal saphenous artery. Ranolazine at therapeutically effective dose significantly recruited muscle microvasculature by increasing muscle microvascular blood volume (∼2-fold, P < 0.05) and increased insulin-mediated whole body glucose disposal (∼30%, P = 0.02). These were associated with an increased insulin delivery into the muscle (P < 0.04). In cultured endothelial cells, ranolazine increased eNOS phosphorylation and cAMP production without affecting endothelial insulin uptake. In ex vivo studies, ranolazine exerted a potent vasodilatatory effect on phenylephrine pre-constricted arterial rings, which was partially abolished by endothelium denudement. In conclusion, ranolazine treatment vasodilatates pre-capillary arterioles and increases microvascular perfusion, which are partially mediated by endothelium, leading to expanded microvascular endothelial surface area available for nutrient and hormone exchanges and resulting in increased muscle delivery and action of insulin. Whether these actions contribute to improved glycaemic control in patients with insulin resistance warrants further investigation.
Collapse
Affiliation(s)
- Zhuo Fu
- Z. Liu: Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, PO Box 801410, Charlottesville, VA 22908, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Escott GM, Jacobus AP, Loss ES. PI3K-dependent actions of insulin and IGF-I on seminiferous tubules from immature rats. Pflugers Arch 2013; 465:1497-505. [DOI: 10.1007/s00424-013-1287-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 04/09/2013] [Accepted: 04/19/2013] [Indexed: 12/21/2022]
|
43
|
Pavlov TS, Ilatovskaya DV, Levchenko V, Li L, Ecelbarger CM, Staruschenko A. Regulation of ENaC in mice lacking renal insulin receptors in the collecting duct. FASEB J 2013; 27:2723-32. [PMID: 23558339 DOI: 10.1096/fj.12-223792] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The epithelial sodium channel (ENaC) is one of the central effectors involved in regulation of salt and water homeostasis in the kidney. To study mechanisms of ENaC regulation, we generated knockout mice lacking the insulin receptor (InsR KO) specifically in the collecting duct principal cells. Single-channel analysis in freshly isolated split-open tubules demonstrated that the InsR-KO mice have significantly lower ENaC activity compared to their wild-type (C57BL/6J) littermates when animals were fed either normal or sodium-deficient diets. Immunohistochemical and Western blot assays demonstrated no significant changes in expression of ENaC subunits in InsR-KO mice compared to wild-type littermates. Insulin treatment caused greater ENaC activity in split-open tubules isolated from wild-type mice but did not have this effect in the InsR-KO mice. Thus, these results suggest that insulin increases ENaC activity via its own receptor affecting the channel open probability. To further determine the mechanism of the action of insulin on ENaC, we used mouse mpkCCDc14 principal cells. Insulin significantly augmented amiloride-sensitive transepithelial flux in these cells. Pretreatment of the mpkCCDc14 cells with phosphatidylinositol 3-kinase (LY294002; 10 μM) or mTOR (PP242; 100 nM) inhibitors precluded this effect. This study provides new information about the importance of insulin receptors expressed in collecting duct principal cells for ENaC activity.
Collapse
Affiliation(s)
- Tengis S Pavlov
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | | | | | | | | | | |
Collapse
|
44
|
Othman EM, Kreissl MC, Kaiser FR, Arias-Loza PA, Stopper H. Insulin-mediated oxidative stress and DNA damage in LLC-PK1 pig kidney cell line, female rat primary kidney cells, and male ZDF rat kidneys in vivo. Endocrinology 2013; 154:1434-43. [PMID: 23456362 DOI: 10.1210/en.2012-1768] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Hyperinsulinemia, a condition with excessively high insulin blood levels, is related to an increased cancer incidence. Diabetes mellitus is the most common of several diseases accompanied by hyperinsulinemia. Because an elevated kidney cancer risk was reported for diabetic patients, we investigated the induction of genomic damage by insulin in LLC-PK1 pig kidney cells, rat primary kidney cells, and ZDF rat kidneys. Insulin at a concentration of 5nM caused a significant increase in DNA damage in vitro. This was associated with the formation of reactive oxygen species (ROS). In the presence of antioxidants, blockers of the insulin, and IGF-I receptors, and a phosphatidylinositol 3-kinase inhibitor, the insulin-mediated DNA damage was reduced. Phosphorylation of protein kinase B (PKB or AKT) was increased and p53 accumulated. Inhibition of the mitochondrial and nicotinamide adenine dinucleotide phosphatase oxidase-related ROS production reduced the insulin-mediated damage. In primary rat cells, insulin also induced genomic damage. In kidneys from healthy, lean ZDF rats, which were infused with insulin to yield normal or high blood insulin levels, while keeping blood glucose levels constant, the amounts of ROS and the tumor protein (p53) were elevated in the high-insulin group compared with the control level group. ROS and p53 were also elevated in diabetic obese ZDF rats. Overall, insulin-induced oxidative stress resulted in genomic damage. If the same mechanisms are active in patients, hyperinsulinemia might cause genomic damage through the induction of ROS contributing to the increased cancer risk, against which the use of antioxidants and/or ROS production inhibitors might exert protective effects.
Collapse
Affiliation(s)
- Eman Maher Othman
- Institute of Pharmacology and Toxicology, University of Würzburg, D-97078 Würzburg, Germany
| | | | | | | | | |
Collapse
|
45
|
Zhao L, Chai W, Fu Z, Dong Z, Aylor KW, Barrett EJ, Cao W, Liu Z. Globular adiponectin enhances muscle insulin action via microvascular recruitment and increased insulin delivery. Circ Res 2013; 112:1263-71. [PMID: 23459195 DOI: 10.1161/circresaha.111.300388] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
RATIONALE Adiponectin enhances insulin action and induces nitric oxide-dependent vasodilatation. Insulin delivery to muscle microcirculation and transendothelial transport are 2 discrete steps that limit insulin's action. We have shown that expansion of muscle microvascular surface area increases muscle insulin delivery and action. OBJECTIVE To examine whether adiponectin modulates muscle microvascular recruitment thus insulin delivery and action in vivo. METHODS AND RESULTS Overnight fasted adult male rats were studied. We determined the effects of adiponectin on muscle microvascular recruitment, using contrast-enhanced ultrasound, on insulin-mediated microvascular recruitment and whole-body glucose disposal, using contrast-enhanced ultrasound and insulin clamp, and on muscle insulin clearance and uptake with (125)I-insulin. Globular adiponectin potently increased muscle microvascular blood volume without altering microvascular blood flow velocity, leading to a significantly increased microvascular blood flow. This was paralleled by a ≈30% to 40% increase in muscle insulin uptake and clearance, and ≈30% increase in insulin-stimulated whole-body glucose disposal. Inhibition of endothelial nitric oxide synthase abolished globular adiponectin-mediated muscle microvascular recruitment and insulin uptake. In cultured endothelial cells, globular adiponectin dose-dependently increased endothelial nitric oxide synthase phosphorylation but had no effect on endothelial cell internalization of insulin. CONCLUSIONS Globular adiponectin increases muscle insulin uptake by recruiting muscle microvasculature, which contributes to its insulin-sensitizing action.
Collapse
Affiliation(s)
- Lina Zhao
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Affiliation(s)
- Zhenqi Liu
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908, USA.
| |
Collapse
|
47
|
Abstract
Vascular endothelium is an important insulin target and plays a pivotal role in the development of metabolic insulin resistance provoked by the Western lifestyle. It acts as a "first-responder" to environmental stimuli such as nutrients, cytokines, chemokines and physical activity and regulates insulin delivery to muscle and adipose tissue and thereby affecting insulin-mediated glucose disposal by these tissues. In addition, it also regulates the delivery of insulin and other appetite regulating signals from peripheral tissues to the central nervous system thus influencing the activity of nuclei that regulate hepatic glucose production, adipose tissue lipolysis and lipogenesis, as well as food consumption. Resistance to insulin's vascular actions therefore broadly impacts tissue function and contribute to metabolic dysregulation. Moreover, vascular insulin resistance negatively impacts vascular health by affecting blood pressure regulation, vessel wall inflammation and atherogenesis thereby contributing to the burden of vascular disease seen with diabetes and metabolic syndrome. In the current review, we examined the evidence that supports the general concept of vascular endothelium as a target of insulin action and discussed the biochemical and physiological consequences of vascular insulin resistance.
Collapse
Affiliation(s)
- Eugene J Barrett
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA.
| | | |
Collapse
|
48
|
Eringa EC, Serne EH, Meijer RI, Schalkwijk CG, Houben AJHM, Stehouwer CDA, Smulders YM, van Hinsbergh VWM. Endothelial dysfunction in (pre)diabetes: characteristics, causative mechanisms and pathogenic role in type 2 diabetes. Rev Endocr Metab Disord 2013; 14:39-48. [PMID: 23417760 DOI: 10.1007/s11154-013-9239-7] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Endothelial dysfunction associated with diabetes and cardiovascular disease is characterized by changes in vasoregulation, enhanced generation of reactive oxygen intermediates, inflammatory activation, and altered barrier function. These endothelial alterations contribute to excess cardiovascular disease in diabetes, but may also play a role in the pathogenesis of diabetes, especially type 2. The mechanisms underlying endothelial dysfunction in diabetes differ between type 1 (T1D) and type 2 diabetes (T2D): hyperglycemia contributes to endothelial dysfunction in all individuals with diabetes, whereas the causative mechanisms in T2D also include impaired insulin signaling in endothelial cells, dyslipidemia and altered secretion of bioactive substances (adipokines) by adipose tissue. The close association of so-called perivascular adipose tissue with arteries and arterioles facilitates the exposure of vascular endothelium to adipokines, particularly if inflammation activates the adipose tissue. Glucose and adipokines activate specific intracellular signaling pathways in endothelium, which in concert result in endothelial dysfunction in diabetes. Here, we review the characteristics of endothelial dysfunction in diabetes, the causative mechanisms involved and the role of endothelial dysfunction(s) in the pathogenesis of T2D. Finally, we will discuss the therapeutic potential of endothelial dysfunction in T2D.
Collapse
Affiliation(s)
- Etto C Eringa
- Departments of Physiology, VU University Medical Center, Amsterdam, the Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
The IGF-1 receptor and regulation of nitric oxide bioavailability and insulin signalling in the endothelium. Pflugers Arch 2013; 465:1065-74. [PMID: 23338941 DOI: 10.1007/s00424-013-1218-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 12/19/2012] [Accepted: 01/07/2013] [Indexed: 12/21/2022]
Abstract
The insulin-like growth factor-1 receptor (IGF-1R), like the insulin receptor (IR), plays a significant role in determining bioavailability of the critical signalling molecule nitric oxide (NO) and hence, modulates endothelial cell function, particularly in response to stimulation with insulin. In particular, the ability of the IGF-1R to form hybrid receptors with the IR appears to be highly significant in determining the sensitivity of the endothelial cell to insulin. This review will examine the structure of the IGF-1R and how this, with particular reference to the ability of the IGF-1R and the IR to form hybrid receptors, may have an effect both on endothelial cell function and the development of cardiovascular disease.
Collapse
|
50
|
Dong Z, Chai W, Wang W, Zhao L, Fu Z, Cao W, Liu Z. Protein kinase A mediates glucagon-like peptide 1-induced nitric oxide production and muscle microvascular recruitment. Am J Physiol Endocrinol Metab 2013; 304. [PMID: 23193054 PMCID: PMC3543568 DOI: 10.1152/ajpendo.00473.2012] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Glucagon-like peptide-1 (GLP-1) causes vasodilation and increases muscle glucose uptake independent of insulin. Recently, we have shown that GLP-1 recruits muscle microvasculature and increases muscle glucose use via a nitric oxide (NO)-dependent mechanism. Protein kinase A (PKA) is a major signaling intermediate downstream of GLP-1 receptors. To examine whether PKA mediates GLP-1's microvascular action in muscle, GLP-1 was infused to overnight-fasted male rats for 120 min in the presence or absence of H89, a PKA inhibitor. Hindleg muscle microvascular recruitment and glucose use were determined. GLP-1 infusion acutely increased muscle microvascular blood volume within 30 min without altering microvascular blood flow velocity or blood pressure. This effect persisted throughout the 120-min infusion period, leading to a significant increase in muscle microvascular blood flow. These changes were paralleled with an approximately twofold increase in plasma NO levels and hindleg glucose extraction. Systemic infusion of H89 completely blocked GLP-1-mediated muscle microvascular recruitment and increases in NO production and muscle glucose extraction. In cultured endothelial cells, GLP-1 acutely increased PKA activity and stimulated endothelial NO synthase phosphorylation at Ser(1177) and NO production. PKA inhibition abolished these effects. In ex vivo studies, perfusion of the distal saphenous artery with GLP-1 induced significant vasorelaxation that was also abolished by pretreatment of the vessels with PKA inhibitor H89. We conclude that GLP-1 recruits muscle microvasculature by expanding microvascular volume and increases glucose extraction in muscle via a PKA/NO-dependent pathway in the vascular endothelium. This may contribute to postprandial glycemic control and complication prevention in diabetes.
Collapse
Affiliation(s)
- Zhenhua Dong
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | |
Collapse
|