1
|
Galifi CA, Dikdan RJ, Kantak D, Bulatowicz JJ, Maingrette K, Gunderson SI, Wood TL. Identifying antisense oligonucleotides for targeted inhibition of insulin receptor isoform A. Front Oncol 2025; 15:1563985. [PMID: 40303997 PMCID: PMC12038057 DOI: 10.3389/fonc.2025.1563985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/10/2025] [Indexed: 05/02/2025] Open
Abstract
Introduction The insulin receptor (IR) is alternatively spliced into two isoforms, IR-A and IR-B. IR-B is primarily associated with metabolic signaling, whereas IR-A is highly expressed during embryogenesis. IR-A specifically has been associated with several aggressive cancers; however, selective targeting of IR-A has proven difficult due to its homology with IR-B. Methods We generated several antisense oligonucleotides (ASOs) that target the exon 10-12 splice junction site present in IR-A, but not IR-B, mRNA. To test the efficacy of the ASOs, we performed lipofectamine transfections of MDA-MB-231 breast cancer, 22Rv1 prostate carcinoma, and Hs822.T Ewing sarcoma cell lines. We also incubated the MDA-MB-231 cell line with the ASOs in the absence of lipofectamine to determine if they are taken into cells unassisted. Results One ASO variant selectively reduced IR-A mRNA levels with minimal impact on IR-B mRNA and significantly reduced total IR protein. The IR-A ASO successfully induced selective IR-A knockdown in MDA-MB-231 breast cancer cells, which was maintained after a one-week incubation with the ASO. The ASO selectively reduced IR-A mRNA when administered to cells in high doses without the use of a vehicle (i.e. gymnotic delivery). The ASO was also effective at reducing IR-A mRNA in Hs822.T Ewing Sarcoma and 22Rv1 prostate carcinoma cells. Discussion We have developed an ASO that targets IR-A with minimal off-target knockdown of IR-B. We hypothesize that the IR-A ASO will be a useful research tool and may have therapeutic value by inhibiting the oncogenic functions of IR-A in cancer cells.
Collapse
Affiliation(s)
- Christopher A. Galifi
- Department of Pharmacology, Physiology, & Neuroscience, Center for Cell Signaling and Cancer Institute of New Jersey, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
| | - Ryan J. Dikdan
- Public Health Research Institute, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
| | - Divyangi Kantak
- Department of Pharmacology, Physiology, & Neuroscience, Center for Cell Signaling and Cancer Institute of New Jersey, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
| | - Joseph J. Bulatowicz
- Department of Pharmacology, Physiology, & Neuroscience, Center for Cell Signaling and Cancer Institute of New Jersey, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
| | - Krystopher Maingrette
- Department of Pharmacology, Physiology, & Neuroscience, Center for Cell Signaling and Cancer Institute of New Jersey, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
| | - Samuel I. Gunderson
- Department of Molecular Biology and Biochemistry, Nelson Biological Laboratories, Rutgers University, Piscataway, NJ, United States
| | - Teresa L. Wood
- Department of Pharmacology, Physiology, & Neuroscience, Center for Cell Signaling and Cancer Institute of New Jersey, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
| |
Collapse
|
2
|
Drozd M, Bruns AF, Yuldasheva NY, Maqbool A, Viswambharan H, Skromna A, Makava N, Cheng CW, Sukumar P, Eades L, Walker AMN, Griffin KJ, Galloway S, Watt NT, Haywood N, Palin V, Warmke N, Imrie H, Bridge K, Beech DJ, Wheatcroft SB, Kearney MT, Cubbon RM. Endothelial insulin-like growth factor-1 signaling regulates vascular barrier function and atherogenesis. Cardiovasc Res 2025:cvaf055. [PMID: 40171617 DOI: 10.1093/cvr/cvaf055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 09/19/2024] [Accepted: 02/15/2025] [Indexed: 04/04/2025] Open
Abstract
AIMS Progressive deposition of cholesterol in the arterial wall characterizes atherosclerosis, which underpins most cases of myocardial infarction and stroke. Insulin-like growth factor-1 (IGF-1) is a hormone that regulates systemic growth and metabolism and possesses anti-atherosclerotic properties. We asked whether endothelial-restricted augmentation of IGF-1 signaling is sufficient to suppress atherogenesis. METHODS AND RESULTS We generated mice with endothelial-restricted over-expression of human wildtype IGF-1R (hIGFREO/ApoE-/-) or a signaling defective K1003R mutant human IGF-1R (mIGFREO/ApoE-/-) and compared them to their respective ApoE-/- littermates. hIGFREO/ApoE-/- had less atherosclerosis, circulating leukocytes, arterial cholesterol uptake, and vascular leakage in multiple organs, whereas mIGFREO/ApoE-/- did not exhibit these phenomena. Overexpressing wildtype IGF-1R in human umbilical vein endothelial cells (HUVEC) altered the localization of tight junction proteins and reduced paracellular leakage across their monolayers, whilst overexpression of K1003R IGF-1R did not have these effects. Moreover, only overexpression of wildtype IGF-1R reduced HUVEC internalization of cholesterol-rich low density lipoprotein particles and increased their association of these particles with clathrin, but not caveolin-1, implicating it in vesicular uptake of lipoproteins. Endothelial overexpression of wildtype versus K1003R IGF-1R also reduced expression of YAP/TAZ target genes and nuclear localization of TAZ, which may be relevant to its impact on vascular barrier and atherogenesis. CONCLUSIONS Endothelial IGF-1 signaling modulates both para- and trans-cellular vascular barrier function. Beyond reducing atherosclerosis, this could have relevance to many diseases associated with abnormal vascular permeability.
Collapse
Affiliation(s)
- Michael Drozd
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Alexander-Francisco Bruns
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Nadira Y Yuldasheva
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Azhar Maqbool
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Hema Viswambharan
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Anna Skromna
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Natallia Makava
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Chew W Cheng
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Piruthivi Sukumar
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Lauren Eades
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Andrew M N Walker
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Kathryn J Griffin
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Stacey Galloway
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Nicole T Watt
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Natalie Haywood
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Victoria Palin
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Nele Warmke
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Helen Imrie
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Katherine Bridge
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - David J Beech
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Stephen B Wheatcroft
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Mark T Kearney
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Richard M Cubbon
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds, LS2 9JT, United Kingdom
| |
Collapse
|
3
|
Luk C, Bridge KI, Warmke N, Simmons KJ, Drozd M, Moran A, MacCannell ADV, Cheng CW, Straw S, Scragg JL, Smith J, Ozber CH, Wilkinson CG, Skromna A, Makava N, Prag HA, Simon Futers T, Brown OI, Bruns AF, Walker AM, Watt NT, Mughal R, Griffin KJ, Yuldasheva NY, Limumpornpetch S, Viswambharan H, Sukumar P, Beech DJ, Vidal-Puig A, Witte KK, Murphy MP, Hartley RC, Wheatcroft SB, Cubbon RM, Roberts LD, Kearney MT, Haywood NJ. Paracrine role of endothelial IGF-1 receptor in depot-specific adipose tissue adaptation in male mice. Nat Commun 2025; 16:170. [PMID: 39747815 PMCID: PMC11696296 DOI: 10.1038/s41467-024-54669-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 11/18/2024] [Indexed: 01/04/2025] Open
Abstract
During recent decades, changes in lifestyle have led to widespread nutritional obesity and its related complications. Remodelling adipose tissue as a therapeutic goal for obesity and its complications has attracted much attention and continues to be actively explored. The endothelium lines all blood vessels and is close to all cells, including adipocytes. The endothelium has been suggested to act as a paracrine organ. We explore the role of endothelial insulin-like growth factor-1 receptor (IGF-1R), as a paracrine modulator of white adipose phenotype. We show that a reduction in endothelial IGF-1R expression in the presence of high-fat feeding in male mice leads to depot-specific beneficial white adipose tissue remodelling, increases whole-body energy expenditure and enhances insulin sensitivity via a non-cell-autonomous paracrine mechanism. We demonstrate that increased endothelial malonate may be contributory and that malonate prodrugs have potentially therapeutically relevant properties in the treatment of obesity-related metabolic disease.
Collapse
Affiliation(s)
- Cheukyau Luk
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Katherine I Bridge
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Nele Warmke
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Katie J Simmons
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- School of Biomedical Sciences, Faculty of Biological Sciences & Astbury Centre, University of Leeds, Leeds, UK
| | - Michael Drozd
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Amy Moran
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Amanda D V MacCannell
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Chew W Cheng
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Sam Straw
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Jason L Scragg
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Jessica Smith
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Claire H Ozber
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- Division of Gastroenterology & Surgery, Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Chloe G Wilkinson
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- North West Genomic Laboratory Hub, Manchester University NHS Foundation Trust, St Mary's Hospital, Oxford Road, Manchester, UK
| | - Anna Skromna
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Natallia Makava
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Hiran A Prag
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - T Simon Futers
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Oliver I Brown
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Alexander-Francisco Bruns
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Andrew Mn Walker
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Nicole T Watt
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Romana Mughal
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- Department of Optometry and Vision Sciences, University of Huddersfield, Huddersfield, UK
| | - Kathryn J Griffin
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Nadira Y Yuldasheva
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Sunti Limumpornpetch
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- Division of Internal Medicine, Cardiology Unit, Faculty of Medicine Prince of Songkla University, Songkhla, Thailand
| | - Hema Viswambharan
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Piruthivi Sukumar
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - David J Beech
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | | | - Klaus K Witte
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | | | - Stephen B Wheatcroft
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Richard M Cubbon
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Lee D Roberts
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Mark T Kearney
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK.
| | - Natalie J Haywood
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| |
Collapse
|
4
|
Myers CG, Viswambharan H, Haywood NJ, Bridge K, Turvey S, Armstrong T, Lunn L, Meakin PJ, Porter KE, Clavane EM, Beech DJ, Cubbon RM, Wheatcroft SB, McPhillie MJ, Issad T, Fishwick CW, Kearney MT, Simmons KJ. Small molecule modulation of insulin receptor-insulin like growth factor-1 receptor heterodimers in human endothelial cells. Mol Cell Endocrinol 2024; 594:112387. [PMID: 39419341 DOI: 10.1016/j.mce.2024.112387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 09/26/2024] [Accepted: 10/12/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVES The insulin receptor (IR) and insulin like growth factor-1 receptor (IGF-1R) are heterodimers consisting of two extracellular α-subunits and two transmembrane β -subunits. Insulin αβ and insulin like growth factor-1 αβ hemi-receptors can heterodimerize to form hybrids composed of one IR αβ and one IGF-1R αβ. The function of hybrids in the endothelium is unclear. We sought insight by developing a small molecule capable of reducing hybrid formation in endothelial cells. METHODS We performed a high-throughput small molecule screening, based on a homology model of the apo hybrid structure. Endothelial cells were studied using western blotting and qPCR to determine the effects of small molecules that reduced hybrid formation. RESULTS Our studies unveil a first-in-class quinoline-containing heterocyclic small molecule that reduces hybrids by >50% in human umbilical vein endothelial cells (HUVECs) with no effects on IR or IGF-1R. This small molecule reduced expression of the negative regulatory p85α subunit of phosphatidylinositol 3-kinase, increased basal phosphorylation of the downstream target Akt and enhanced insulin/insulin-like growth factor-1 and shear stress-induced serine phosphorylation of Akt. In primary saphenous vein endothelial cells (SVEC) from patients with type 2 diabetes mellitus undergoing coronary artery bypass (CABG) surgery, hybrid receptor expression was greater than in patients without type 2 diabetes mellitus. The small molecule significantly reduced hybrid expression in SVEC from patients with type 2 diabetes mellitus. CONCLUSIONS We identified a small molecule that decreases the formation of IR: IGF-1R hybrid receptors in human endothelial cells, without significant impact on the overall expression of IR or IGF-1R. In HUVECs, reduction of IR: IGF-1R hybrid receptors leads to an increase in insulin-induced serine phosphorylation of the critical downstream signalling kinase, Akt. The underpinning mechanism appears, at least in part to involve the attenuation of the inhibitory effect of IR: IGF-1R hybrid receptors on PI3-kinase signalling.
Collapse
Affiliation(s)
- Chloe G Myers
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Hema Viswambharan
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Natalie J Haywood
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Katherine Bridge
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Samuel Turvey
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Tom Armstrong
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Lydia Lunn
- Department of Chemistry University of Leeds, Leeds, United Kingdom
| | - Paul J Meakin
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Karen E Porter
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Eva M Clavane
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - David J Beech
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom; National Institute for Health and Care Research Leeds Biomedical Research Centre, Leeds, United Kingdom
| | - Richard M Cubbon
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom; National Institute for Health and Care Research Leeds Biomedical Research Centre, Leeds, United Kingdom
| | - Stephen B Wheatcroft
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | | | - Tarik Issad
- Université Paris Cité, CNRS, INSERM, Institut Cochin, F-75014, Paris, France
| | | | - Mark T Kearney
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom; National Institute for Health and Care Research Leeds Biomedical Research Centre, Leeds, United Kingdom.
| | - Katie J Simmons
- School of Biomedical Sciences, Faculty of Biological Sciences & Astbury Centre, University of Leeds, United Kingdom
| |
Collapse
|
5
|
Walker AM, Watt NT, Yuldasheva NY, Dalmia S, Conning-Rowland M, Cheng CW, Warmke N, Bridge K, Brown OI, Luk C, Drozd M, Haywood NJ, Skromna A, Makava N, Wheatcroft SB, Kearney MT, Cubbon RM. Distinct, common and synergistic effects of insulin and IGF-1 receptors on healthy murine ageing. Heliyon 2024; 10:e36457. [PMID: 39247377 PMCID: PMC11379992 DOI: 10.1016/j.heliyon.2024.e36457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/29/2024] [Accepted: 08/15/2024] [Indexed: 09/10/2024] Open
Abstract
Objective Reduced IGF-1 signalling is an evolutionarily conserved mediator of longevity, yet the magnitude of this effect is substantially larger in organisms retaining a common insulin and IGF-1 receptor. Whether this reflects the failure to simultaneously reduce IGF-1 and insulin signalling in mammalian model systems remains unexplored, as is the associated impact on markers of healthy ageing. We set out to address these uncertainties. Methods We compared the duration of healthy life (healthspan) in male mice with haploinsufficiency of the insulin receptor (IRKO), IGF-1 receptor (IGF-1RKO), or both (DKO), versus wildtype (WT) littermates. Cognitive performance was defined using nesting studies at 3- and 24-months of age. Brain transcriptome was characterised at 3- and 18-months of age using RNA-seq. Results Healthspan was longer in DKO versus WT, with IRKO and IGF-1RKO being intermediate. At 2 years of age, DKO also exhibited preserved nesting behaviour in contrast with all other genotypes. Differential insulin sensitivity or weight gain during ageing did not explain the preserved healthspan of DKO, since these were comparable to IRKO littermates. Brain transcriptomics at 18 months of age revealed lower expression of canonical ageing-associated genes in DKO versus WT, although many of these findings were replicated in IRKO versus WT or IGF-1RKO vs WT. Conclusions Reduced insulin and IGF-1 receptor expression have both common and synergistic effects upon elements of healthy mammalian ageing, suggesting future ageing studies should consider targeting both insulin and IGF-1 signalling.
Collapse
Affiliation(s)
- Andrew Mn Walker
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| | - Nicole T Watt
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| | - Nadira Y Yuldasheva
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| | - Sanjush Dalmia
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| | - Marcella Conning-Rowland
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| | - Chew W Cheng
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| | - Nele Warmke
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| | - Katherine Bridge
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| | - Oliver I Brown
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| | - Cheukyau Luk
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| | - Michael Drozd
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| | - Natalie J Haywood
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| | - Anna Skromna
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| | - Natasha Makava
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| | - Stephen B Wheatcroft
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| | - Mark T Kearney
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| | - Richard M Cubbon
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| |
Collapse
|
6
|
Turvey S, Muench SP, Issad T, Fishwick CWG, Kearney MT, Simmons KJ. Using site-directed mutagenesis to further the understanding of insulin receptor-insulin like growth factor-1 receptor heterodimer structure. Growth Horm IGF Res 2024; 77:101607. [PMID: 39033666 DOI: 10.1016/j.ghir.2024.101607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/26/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Type 2 diabetes is characterised by the disruption of insulin and insulin-like growth factor (IGF) signalling. The key hubs of these signalling cascades - the Insulin receptor (IR) and Insulin-like growth factor 1 receptor (IGF1R) - are known to form functional IR-IGF1R hybrid receptors which are insulin resistant. However, the mechanisms underpinning IR-IGF1R hybrid formation are not fully understood, hindering the ability to modulate this for future therapies targeting this receptor. To pinpoint suitable sites for intervention, computational hotspot prediction was utilised to identify promising epitopes for targeting with point mutagenesis. Specific IGF1R point mutations F450A, R391A and D555A show reduced affinity of the hybrid receptor in a BRET based donor-saturation assay, confirming hybrid formation could be modulated at this interface. These data provide the basis for rational design of more effective hybrid receptor modulators, supporting the prospect of identifying a small molecule that specifically interacts with this target.
Collapse
Affiliation(s)
- Samuel Turvey
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Stephen P Muench
- School of Biomedical Sciences, Faculty of Biological Sciences & Astbury Centre, University of Leeds, UK
| | - Tarik Issad
- Université Paris Cité, CNRS, INSERM, Institut Cochin, F-75014 Paris, France
| | | | - Mark T Kearney
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Katie J Simmons
- School of Biomedical Sciences, Faculty of Biological Sciences & Astbury Centre, University of Leeds, UK.
| |
Collapse
|
7
|
Abdellatif M, Rainer PP, Sedej S, Kroemer G. Hallmarks of cardiovascular ageing. Nat Rev Cardiol 2023; 20:754-777. [PMID: 37193857 DOI: 10.1038/s41569-023-00881-3] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/21/2023] [Indexed: 05/18/2023]
Abstract
Normal circulatory function is a key determinant of disease-free life expectancy (healthspan). Indeed, pathologies affecting the cardiovascular system, which are growing in prevalence, are the leading cause of global morbidity, disability and mortality, whereas the maintenance of cardiovascular health is necessary to promote both organismal healthspan and lifespan. Therefore, cardiovascular ageing might precede or even underlie body-wide, age-related health deterioration. In this Review, we posit that eight molecular hallmarks are common denominators in cardiovascular ageing, namely disabled macroautophagy, loss of proteostasis, genomic instability (in particular, clonal haematopoiesis of indeterminate potential), epigenetic alterations, mitochondrial dysfunction, cell senescence, dysregulated neurohormonal signalling and inflammation. We also propose a hierarchical order that distinguishes primary (upstream) from antagonistic and integrative (downstream) hallmarks of cardiovascular ageing. Finally, we discuss how targeting each of the eight hallmarks might be therapeutically exploited to attenuate residual cardiovascular risk in older individuals.
Collapse
Affiliation(s)
- Mahmoud Abdellatif
- Department of Cardiology, Medical University of Graz, Graz, Austria.
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- BioTechMed Graz, Graz, Austria.
| | - Peter P Rainer
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| |
Collapse
|
8
|
Luca AC, David SG, David AG, Țarcă V, Pădureț IA, Mîndru DE, Roșu ST, Roșu EV, Adumitrăchioaiei H, Bernic J, Cojocaru E, Țarcă E. Atherosclerosis from Newborn to Adult-Epidemiology, Pathological Aspects, and Risk Factors. Life (Basel) 2023; 13:2056. [PMID: 37895437 PMCID: PMC10608492 DOI: 10.3390/life13102056] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/02/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Cardiovascular disease is the leading cause of mortality and morbidity throughout the world, accounting for 16.7 million deaths each year. The underlying pathological process for the majority of cardiovascular diseases is atherosclerosis, a slowly progressing, multifocal, chronic, immune-inflammatory disease that involves the intima of large and medium-sized arteries. The process of atherosclerosis begins in childhood as fatty streaks-an accumulation of lipids, inflammatory cells, and smooth muscle cells in the arterial wall. Over time, a more complex lesion develops into an atheroma and characteristic fibrous plaques. Atherosclerosis alone is rarely fatal; it is the further changes that render fibrous plaques vulnerable to rupture; plaque rupture represents the most common cause of coronary thrombosis. The prevalence of atherosclerosis is increasing worldwide and more than 50% of people with circulatory disease die of it, mostly in modern societies. Epidemiological studies have revealed several environmental and genetic risk factors that are associated with the early formation of a pathogenic foundation for atherosclerosis, such as dyslipidemia, hypertension, diabetes mellitus, obesity, and smoking. The purpose of this review is to bring together the current information concerning the origin and progression of atherosclerosis in childhood as well as the identification of known risk factors for atherosclerotic cardiovascular disease in children.
Collapse
Affiliation(s)
- Alina Costina Luca
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.C.L.); (D.E.M.); (E.V.R.)
| | - Simona Georgiana David
- Saint Mary Emergency Hospital for Children, 700309 Iasi, Romania; (S.G.D.); (A.G.D.); (I.-A.P.); (H.A.)
| | - Alexandru Gabriel David
- Saint Mary Emergency Hospital for Children, 700309 Iasi, Romania; (S.G.D.); (A.G.D.); (I.-A.P.); (H.A.)
| | - Viorel Țarcă
- Department of Preventive Medicine and Interdisciplinarity, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Ioana-Alexandra Pădureț
- Saint Mary Emergency Hospital for Children, 700309 Iasi, Romania; (S.G.D.); (A.G.D.); (I.-A.P.); (H.A.)
| | - Dana Elena Mîndru
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.C.L.); (D.E.M.); (E.V.R.)
| | - Solange Tamara Roșu
- Nursing Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Eduard Vasile Roșu
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.C.L.); (D.E.M.); (E.V.R.)
| | - Heidrun Adumitrăchioaiei
- Saint Mary Emergency Hospital for Children, 700309 Iasi, Romania; (S.G.D.); (A.G.D.); (I.-A.P.); (H.A.)
| | - Jana Bernic
- Discipline of Pediatric Surgery, “Nicolae Testemițanu” State University of Medicine and Pharmacy, 2025 Chisinau, Moldova;
| | - Elena Cojocaru
- Department of Morphofunctional Sciences I—Pathology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Elena Țarcă
- Surgery II Department—Pediatric Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| |
Collapse
|
9
|
Xiong Y, Wang Y, Yang T, Luo Y, Xu S, Li L. Receptor Tyrosine Kinase: Still an Interesting Target to Inhibit the Proliferation of Vascular Smooth Muscle Cells. Am J Cardiovasc Drugs 2023; 23:497-518. [PMID: 37524956 DOI: 10.1007/s40256-023-00596-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/05/2023] [Indexed: 08/02/2023]
Abstract
Vascular smooth muscle cells (VSMCs) proliferation is a critical event that contributes to the pathogenesis of vascular remodeling such as hypertension, restenosis, and pulmonary hypertension. Increasing evidences have revealed that VSMCs proliferation is associated with the activation of receptor tyrosine kinases (RTKs) by their ligands, including the insulin-like growth factor receptor (IGFR), fibroblast growth factor receptor (FGFR), epidermal growth factor receptor (EGFR), vascular endothelial growth factor receptor (VEGFR), and platelet-derived growth factor receptor (PDGFR). Moreover, some receptor tyrosinase inhibitors (TKIs) have been found and can prevent VSMCs proliferation to attenuate vascular remodeling. Therefore, this review will describe recent research progress on the role of RTKs and their inhibitors in controlling VSMCs proliferation, which helps to better understand the function of VSMCs proliferation in cardiovascular events and is beneficial for the prevention and treatment of vascular disease.
Collapse
Affiliation(s)
- Yilin Xiong
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China
- Department of Pharmacology, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Zunyi, 563000, Guizhou, China
| | - Yan Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China
- Department of Pharmacology, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Zunyi, 563000, Guizhou, China
| | - Tao Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China
- Department of Pharmacology, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Zunyi, 563000, Guizhou, China
| | - Yunmei Luo
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China
- Department of Pharmacology, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Zunyi, 563000, Guizhou, China
| | - Shangfu Xu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China
- Department of Pharmacology, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Zunyi, 563000, Guizhou, China
| | - Lisheng Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China.
- Department of Pharmacology, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Zunyi, 563000, Guizhou, China.
| |
Collapse
|
10
|
Imrie H, Viswambharan H, Haywood NJ, Bridge KI, Yuldasheva NY, Galloway S, Simmons KJ, Cubbon RM, Sukumar P, Watt NT, Lichtenstein L, Wyatt JI, Kudo H, Goldin R, Rode B, Wheatcroft SB, Kearney MT. Cixutumumab reveals a critical role for IGF-1 in adipose and hepatic tissue remodelling during the development of diet-induced obesity. Adipocyte 2022; 11:366-378. [PMID: 35734881 PMCID: PMC9235901 DOI: 10.1080/21623945.2022.2089394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
High fat diet (HFD)-induced obesity leads to perturbation in the storage function of white adipose tissue (WAT) resulting in deposition of lipids in tissues ill-equipped to deal with this challenge. The role of insulin like growth factor-1 (IGF-1) in the systemic and organ-specific responses to HFD is unclear. Using cixutumumab, a monoclonal antibody that internalizes and degrades cell surface IGF-1 receptors (IGF-1 R), leaving insulin receptor expression unchanged we aimed to establish the role of IGF-1 R in the response to a HFD. Mice treated with cixutumumab fed standard chow developed mild hyperinsulinemia with no change in WAT. When challenged by HFD mice treated with cixutumumab had reduced weight gain, reduced WAT expansion, and reduced hepatic lipid vacuole formation. In HFD-fed mice, cixutumumab led to reduced levels of genes encoding proteins important in fatty acid metabolism in WAT and liver. Cixutumumab protected against blunting of insulin-stimulated phosphorylation of Akt in liver of HFD fed mice. These data reveal an important role for IGF-1 R in the WAT and hepatic response to short-term nutrient excess. IGF-1 R inhibition during HFD leads to a lipodystrophic phenotype with a failure of WAT lipid storage and protection from HFD-induced hepatic insulin resistance.
Collapse
Affiliation(s)
- Helen Imrie
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Hema Viswambharan
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Natalie J Haywood
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Katherine I Bridge
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Nadira Y Yuldasheva
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Stacey Galloway
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Katie J Simmons
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Richard M Cubbon
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Piruthivi Sukumar
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Nicole T Watt
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Laeticia Lichtenstein
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Judy I Wyatt
- Department of Pathology, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Hiromi Kudo
- Department of Metabolism, Digestion and Reproduction, Imperial College, London, United Kingdom
| | - Robert Goldin
- Department of Metabolism, Digestion and Reproduction, Imperial College, London, United Kingdom
| | - Baptiste Rode
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Stephen B Wheatcroft
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Mark T Kearney
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
11
|
Kopchick JJ, Basu R, Berryman DE, Jorgensen JOL, Johannsson G, Puri V. Covert actions of growth hormone: fibrosis, cardiovascular diseases and cancer. Nat Rev Endocrinol 2022; 18:558-573. [PMID: 35750929 PMCID: PMC9703363 DOI: 10.1038/s41574-022-00702-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/19/2022] [Indexed: 12/20/2022]
Abstract
Since its discovery nearly a century ago, over 100,000 studies of growth hormone (GH) have investigated its structure, how it interacts with the GH receptor and its multiple actions. These include effects on growth, substrate metabolism, body composition, bone mineral density, the cardiovascular system and brain function, among many others. Recombinant human GH is approved for use to promote growth in children with GH deficiency (GHD), along with several additional clinical indications. Studies of humans and animals with altered levels of GH, from complete or partial GHD to GH excess, have revealed several covert or hidden actions of GH, such as effects on fibrosis, cardiovascular function and cancer. In this Review, we do not concentrate on the classic and controversial indications for GH therapy, nor do we cover all covert actions of GH. Instead, we stress the importance of the relationship between GH and fibrosis, and how fibrosis (or lack thereof) might be an emerging factor in both cardiovascular and cancer pathologies. We highlight clinical data from patients with acromegaly or GHD, alongside data from cellular and animal studies, to reveal novel phenotypes and molecular pathways responsible for these actions of GH in fibrosis, cardiovascular function and cancer.
Collapse
Affiliation(s)
- John J Kopchick
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA.
- The Diabetes Institute, Ohio University, Athens, OH, USA.
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA.
| | - Reetobrata Basu
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- The Diabetes Institute, Ohio University, Athens, OH, USA
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
| | - Darlene E Berryman
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- The Diabetes Institute, Ohio University, Athens, OH, USA
| | - Jens O L Jorgensen
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Gudmundur Johannsson
- Department of Endocrinology, Sahlgrenska University Hospital, Sahlgrenska Academy, University of Göteborg, Gothenburg, Sweden
| | - Vishwajeet Puri
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- The Diabetes Institute, Ohio University, Athens, OH, USA
| |
Collapse
|
12
|
Powell MJ, Fuller S, Gunderson EP, Benz CC. Reduced cardiovascular risks in women with endometriosis or polycystic ovary syndrome carrying a common functional IGF1R variant. Hum Reprod 2022; 37:1083-1094. [PMID: 35362533 PMCID: PMC9071223 DOI: 10.1093/humrep/deac059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 03/03/2022] [Indexed: 11/28/2022] Open
Abstract
STUDY QUESTION Is the increased future cardiovascular risk seen in women with endometriosis or polycystic ovary syndrome (PCOS) mitigated by functional insulin-like growth factor-1 receptor (IGF1R) single-nucleotide polymorphism (SNP) rs2016347 as previously shown in women with hypertensive disorders of pregnancy? SUMMARY ANSWER This cohort study found that women with endometriosis or PCOS who carry a T allele of IGF1R SNP rs2016347 had a reduced future risk of developing cardiovascular disease (CVD) and associated risk factors, with risk reduction dependent on cohort era. WHAT IS KNOWN ALREADY Women with endometriosis or PCOS have been shown to have an increased future risk of CVD and associated risk factors with limited predictive ability. STUDY DESIGN, SIZE, DURATION This retrospective cohort study took place in the Nurses' Health Study 2 (NHS2), which enrolled 116 430 participants in 1989 who were followed through 2015. The study population was analyzed in its entirety, and subdivided into entry (pre-1989) and after entry (post-1989) exposure cohorts. All NHS2 participants were eligible for inclusion in the study, 9599 (8.2%) were excluded for missing covariates. PARTICIPANTS/MATERIALS, SETTING, METHODS The NHS2 enrolled female registered nurses from 14 different states who ranged in age from 25 to 42 years at study entry. Data were collected from entry and biennial questionnaires, and analysis conducted from November 2020 to June 2021. Cox proportional hazard models were used to assess risk of CVD, hypertension (HTN), hypercholesterolemia (HC) and type 2 diabetes, both with and without genotyping for rs2016347. MAIN RESULTS AND THE ROLE OF CHANCE While women without endometriosis or PCOS, as a whole, demonstrated no impact of genotype on risk in either cohort, women with endometriosis carrying a T allele had a lower risk of CVD (hazard ratio (HR), 0.48; 95% CI, 0.27-0.86, P = 0.02) and HTN (HR, 0.80; 95% CI, 0.66-0.97, P = 0.03) in the pre-1989 cohort, while those in the post-1989 cohort had a decrease in risk for HC (HR, 0.76; 95% CI, 0.62-0.94, P = 0.01). Women with PCOS in the post-1989 cohort showed a significant protective impact of the T allele on HTN (HR, 0.44; 95% CI, 0.27-0.73, P = 0.002) and HC (HR, 0.62; 95% CI, 0.40-0.95, P = 0.03). LIMITATIONS, REASONS FOR CAUTION Data on specific endometriosis lesion locations or disease stage, as well as on PCOS phenotypes were lacking. In addition, data on systemic medical treatments beyond the use of oral contraceptives were missing, and these treatments may have confounded the results. WIDER IMPLICATIONS OF THE FINDINGS These findings implicate systemic dysregulation of the insulin-like growth factor-1 axis in the development of HTN, HC and clinical CVD in endometriosis and PCOS, suggesting a common underlying pathogenetic mechanism. STUDY FUNDING/COMPETING INTEREST(S) The NHS2 infrastructure for questionnaire data collection was supported by National Institute of Health (NIH) grant U01CA176726. This work was also supported in part by NIH and National Cancer Institute grant U24CA210990; as well, research effort and publication costs were supported by the Elizabeth MA Stevens donor funds provided to the Buck Institute for Research on Aging. The authors declare they have no conflicts of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Mark J Powell
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Sophia Fuller
- Graduate Group in Biostatistics, University of California, Berkeley, School of Public Health, Berkeley, CA, USA
| | - Erica P Gunderson
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
- Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA, USA
| | | |
Collapse
|
13
|
Turvey SJ, McPhillie MJ, Kearney MT, Muench SP, Simmons KJ, Fishwick CWG. Recent developments in the structural characterisation of the IR and IGF1R: implications for the design of IR-IGF1R hybrid receptor modulators. RSC Med Chem 2022; 13:360-374. [PMID: 35647546 PMCID: PMC9020618 DOI: 10.1039/d1md00300c] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 01/12/2022] [Indexed: 12/16/2022] Open
Abstract
The insulin receptor (IR) and insulin-like growth factor 1 receptor (IGF1R) are dimeric disulfide-linked receptor tyrosine kinases, whose actions regulate metabolic and mitogenic signalling pathways inside the cell. It is well documented that in tissues co-expressing the IR and IGF1R, their respective monomers can heterodimerise to form IR-IGF1R hybrid receptors. Increased populations of the IR-IGF1R hybrid receptors are associated with several disease states, including type 2 diabetes and cancer. Recently, progress in the structural biology of IR and IGF1R has given insights into their structure-function relationships and mechanism of action. However, challenges in isolating IR-IGF1R hybrid receptors mean that their structural properties remain relatively unexplored. This review discusses the advances in the structural understanding of the IR and IGF1R, and how these discoveries can inform the design of small-molecule modulators of the IR-IGF1R hybrid receptors to understand their role in cell biology.
Collapse
Affiliation(s)
- Samuel J Turvey
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds UK
| | | | - Mark T Kearney
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds UK
| | - Stephen P Muench
- School of Biomedical Sciences, Faculty of Biological Sciences & Astbury Centre, University of Leeds UK
| | - Katie J Simmons
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds UK
| | | |
Collapse
|
14
|
Qian Y, Berryman DE, Basu R, List EO, Okada S, Young JA, Jensen EA, Bell SRC, Kulkarni P, Duran-Ortiz S, Mora-Criollo P, Mathes SC, Brittain AL, Buchman M, Davis E, Funk KR, Bogart J, Ibarra D, Mendez-Gibson I, Slyby J, Terry J, Kopchick JJ. Mice with gene alterations in the GH and IGF family. Pituitary 2022; 25:1-51. [PMID: 34797529 PMCID: PMC8603657 DOI: 10.1007/s11102-021-01191-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/21/2021] [Indexed: 01/04/2023]
Abstract
Much of our understanding of GH's action stems from animal models and the generation and characterization of genetically altered or modified mice. Manipulation of genes in the GH/IGF1 family in animals started in 1982 when the first GH transgenic mice were produced. Since then, multiple laboratories have altered mouse DNA to globally disrupt Gh, Ghr, and other genes upstream or downstream of GH or its receptor. The ability to stay current with the various genetically manipulated mouse lines within the realm of GH/IGF1 research has been daunting. As such, this review attempts to consolidate and summarize the literature related to the initial characterization of many of the known gene-manipulated mice relating to the actions of GH, PRL and IGF1. We have organized the mouse lines by modifications made to constituents of the GH/IGF1 family either upstream or downstream of GHR or to the GHR itself. Available data on the effect of altered gene expression on growth, GH/IGF1 levels, body composition, reproduction, diabetes, metabolism, cancer, and aging are summarized. For the ease of finding this information, key words are highlighted in bold throughout the main text for each mouse line and this information is summarized in Tables 1, 2, 3 and 4. Most importantly, the collective data derived from and reported for these mice have enhanced our understanding of GH action.
Collapse
Affiliation(s)
- Yanrong Qian
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
| | - Darlene E Berryman
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
| | - Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
| | - Shigeru Okada
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Pediatrics, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Jonathan A Young
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Elizabeth A Jensen
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- Translational Biomedical Sciences Doctoral Program, Ohio University, Athens, OH, USA
| | - Stephen R C Bell
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Prateek Kulkarni
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH, USA
- Molecular and Cellular Biology Program, Ohio University, Athens, OH, USA
| | | | - Patricia Mora-Criollo
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Translational Biomedical Sciences Doctoral Program, Ohio University, Athens, OH, USA
| | - Samuel C Mathes
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
| | - Alison L Brittain
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- Molecular and Cellular Biology Program, Ohio University, Athens, OH, USA
| | - Mat Buchman
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Emily Davis
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH, USA
- Molecular and Cellular Biology Program, Ohio University, Athens, OH, USA
| | - Kevin R Funk
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH, USA
- Molecular and Cellular Biology Program, Ohio University, Athens, OH, USA
| | - Jolie Bogart
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH, USA
| | - Diego Ibarra
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Chemistry and Biochemistry, College of Arts and Sciences, Ohio University, Athens, OH, USA
| | - Isaac Mendez-Gibson
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- College of Health Sciences and Professions, Ohio University, Athens, OH, USA
| | - Julie Slyby
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH, USA
| | - Joseph Terry
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH, USA
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA.
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA.
| |
Collapse
|
15
|
Koutouroushis C, Sarkar O. Role of Autophagy in Cardiovascular Disease and Aging. Cureus 2021; 13:e20042. [PMID: 34873555 PMCID: PMC8631374 DOI: 10.7759/cureus.20042] [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] [Accepted: 11/29/2021] [Indexed: 11/15/2022] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide and is expected to further increase as people continue to live even longer. Although the life span of the general population is increasing, the con of such a prolonged life span is that aging has certain detrimental effects on the molecular, structural, and functional elements of the cardiovascular system. This review will discuss various molecular pathways linked to longevity, most notably autophagy and its associated mechanisms, and how these pathways can be targeted to promote cardiovascular health through the process of aging. It is to be noted that the process of autophagy decreases with aging; hence, this review concludes that the promotion of autophagy, through implementation of caloric restriction, intermittent fasting, and pharmacologic agents, has proven to be an efficacious means of stimulating cardiovascular health. Therefore, autophagy is an important target for prevention and procrastination of cardiovascular pathologies in the geriatric population.
Collapse
Affiliation(s)
| | - Oiendrila Sarkar
- General Internal Medicine, St. Mary's Hospital, Isle of Wight NHS Trust, Newport, GBR
| |
Collapse
|
16
|
Tarantini S, Nyúl-Tóth Á, Yabluchanskiy A, Csipo T, Mukli P, Balasubramanian P, Ungvari A, Toth P, Benyo Z, Sonntag WE, Ungvari Z, Csiszar A. Endothelial deficiency of insulin-like growth factor-1 receptor (IGF1R) impairs neurovascular coupling responses in mice, mimicking aspects of the brain aging phenotype. GeroScience 2021; 43:2387-2394. [PMID: 34383203 PMCID: PMC8599783 DOI: 10.1007/s11357-021-00405-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/15/2021] [Indexed: 11/27/2022] Open
Abstract
Age-related impairment of neurovascular coupling (NVC; or "functional hyperemia") compromises moment-to-moment adjustment of regional cerebral blood flow to increased neuronal activity and thereby contributes to the pathogenesis of vascular cognitive impairment (VCI). Previous studies established a causal link among age-related decline in circulating levels of insulin-like growth factor-1 (IGF-1), neurovascular dysfunction and cognitive impairment. Endothelium-mediated microvascular dilation plays a central role in NVC responses. To determine the functional consequences of impaired IGF-1 input to cerebromicrovascular endothelial cells, endothelium-mediated NVC responses were studied in a novel mouse model of accelerated neurovascular aging: mice with endothelium-specific knockout of IGF1R (VE-Cadherin-CreERT2/Igf1rf/f). Increases in cerebral blood flow in the somatosensory whisker barrel cortex (assessed using laser speckle contrast imaging through a cranial window) in response to contralateral whisker stimulation were significantly attenuated in VE-Cadherin-CreERT2/Igf1rf/f mice as compared to control mice. In VE-Cadherin-CreERT2/Igf1rf/f mice, the effects of the NO synthase inhibitor L-NAME were significantly decreased, suggesting that endothelium-specific disruption of IGF1R signaling impairs the endothelial NO-dependent component of NVC responses. Collectively, these findings provide additional evidence that IGF-1 is critical for cerebromicrovascular endothelial health and maintenance of normal NVC responses.
Collapse
Affiliation(s)
- Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731042, USA
- International Training Program in Geroscience, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731042, USA
- Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK, 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Ádám Nyúl-Tóth
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731042, USA
- International Training Program in Geroscience, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731042, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Szeged, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731042, USA
- International Training Program in Geroscience, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731042, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tamas Csipo
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731042, USA
- International Training Program in Geroscience, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731042, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Peter Mukli
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731042, USA
- International Training Program in Geroscience, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731042, USA
- International Training Program in Geroscience, Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Priya Balasubramanian
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731042, USA
| | - Anna Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731042, USA
| | - Peter Toth
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Neurosurgery, University of Pécs Clinical Center, 72359, Pecs, Baranya, Hungary
| | - Zoltan Benyo
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - William E Sonntag
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731042, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731042, USA
- International Training Program in Geroscience, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731042, USA
- Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK, 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731042, USA.
- International Training Program in Geroscience, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731042, USA.
- Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK, 73104, USA.
- Vascular Cognitive Impairment and Neurodegeneration Program, Department of Translational Medicine, Semmelweis University, Budapest, Hungary.
- Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences, Center 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
17
|
Haywood NJ, Luk C, Bridge KI, Drozd M, Makava N, Skromna A, Maccannell A, Ozber CH, Warmke N, Wilkinson CG, Watt NT, Koch‐Paszkowski J, Teh I, Boyle JH, Smart S, Schneider JE, Yuldasheva NY, Roberts LD, Beech DJ, Sukumar P, Wheatcroft SB, Cubbon RM, Kearney MT. Endothelial IGF-1 receptor mediates crosstalk with the gut wall to regulate microbiota in obesity. EMBO Rep 2021; 22:e50767. [PMID: 33934497 PMCID: PMC8097321 DOI: 10.15252/embr.202050767] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 03/12/2021] [Accepted: 03/22/2021] [Indexed: 12/20/2022] Open
Abstract
Changes in composition of the intestinal microbiota are linked to the development of obesity and can lead to endothelial cell (EC) dysfunction. It is unknown whether EC can directly influence the microbiota. Insulin-like growth factor-1 (IGF-1) and its receptor (IGF-1R) are critical for coupling nutritional status and cellular growth; IGF-1R is expressed in multiple cell types including EC. The role of ECIGF-1R in the response to nutritional obesity is unexplored. To examine this, we use gene-modified mice with EC-specific overexpression of human IGF-1R (hIGFREO) and their wild-type littermates. After high-fat feeding, hIGFREO weigh less, have reduced adiposity and have improved glucose tolerance. hIGFREO show an altered gene expression and altered microbial diversity in the gut, including a relative increase in the beneficial genus Akkermansia. The depletion of gut microbiota with broad-spectrum antibiotics induces a loss of the favourable metabolic differences seen in hIGFREO mice. We show that IGF-1R facilitates crosstalk between the EC and the gut wall; this crosstalk protects against diet-induced obesity, as a result of an altered gut microbiota.
Collapse
Affiliation(s)
- Natalie J Haywood
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Cheukyau Luk
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Katherine I Bridge
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Michael Drozd
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Natallia Makava
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Anna Skromna
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Amanda Maccannell
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Claire H Ozber
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Nele Warmke
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Chloe G Wilkinson
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Nicole T Watt
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Joanna Koch‐Paszkowski
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Irvin Teh
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Jordan H Boyle
- Faculty of EngineeringSchool of Mechanical EngineeringUniversity of LeedsLeedsUK
| | - Sean Smart
- Department of OncologyUniversity of OxfordOxfordUK
| | - Jurgen E Schneider
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Nadira Y Yuldasheva
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Lee D Roberts
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - David J Beech
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Piruthivi Sukumar
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Stephen B Wheatcroft
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Richard M Cubbon
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Mark T Kearney
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| |
Collapse
|
18
|
Yan C, Zhao M, Li S, Liu T, Xu C, Liu L, Geng T, Gong D. Increase of E3 ubiquitin ligase NEDD4 expression leads to degradation of its target proteins PTEN/IGF1R during the formation of goose fatty liver. J Anim Sci 2020; 98:5897041. [PMID: 32841331 DOI: 10.1093/jas/skaa270] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/22/2020] [Indexed: 11/12/2022] Open
Abstract
Goose fatty liver may have a unique protective mechanism as it does not show a pathological injury even in the case of severe steatosis. Although neural precursor cell-expressed developmentally downregulated gene 4 (NEDD4) participates in repair and regeneration of injured liver through its target proteins, its role in nonalcoholic fatty liver disease remains unknown. Using quantitative polymerase chain reaction (PCR) and immunoblot analyses, here, we found that the messenger RNA (mRNA) and protein expressions of NEDD4 were induced in goose fatty liver compared with normal liver. The mRNA expression of the gene of phosphate and tension homology deleted on chromosome ten (PTEN) and insulin-like growth factor 1 receptor (IGF1R) was also induced in goose fatty liver; however, their protein expression was or tended to be suppressed. Moreover, the co-immunoprecipitation analysis indicated that there was a physical association between NEDD4 and PTEN in goose liver, which was consistent with the ubiquitination of PTEN in goose fatty liver. Furthermore, NEDD4 overexpression in goose primary hepatocytes suppressed the PTEN and IGF1R protein levels without a significant effect on their mRNA expression. In conclusion, the increased expression of NEDD4 leads to the degradation of PTEN and IGF1R proteins through ubiquitination in goose fatty liver, suggesting that NEDD4 may protect goose fatty liver from severe steatosis-associated injury via its target proteins during the development of goose fatty liver.
Collapse
Affiliation(s)
- Chunchi Yan
- College of Animal Science and Technology, Yangzhou University, Yangzhou, P.R. China
| | - Minmeng Zhao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, P.R. China
| | - Shuo Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou, P.R. China
| | - Tongjun Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, P.R. China
| | - Cheng Xu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, P.R. China
| | - Long Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, P.R. China
| | - Tuoyu Geng
- College of Animal Science and Technology, Yangzhou University, Yangzhou, P.R. China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu Province, P. R. China
| | - Daoqing Gong
- College of Animal Science and Technology, Yangzhou University, Yangzhou, P.R. China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu Province, P. R. China
| |
Collapse
|
19
|
Sidorkiewicz I, Jóźwik M, Niemira M, Krętowski A. Insulin Resistance and Endometrial Cancer: Emerging Role for microRNA. Cancers (Basel) 2020; 12:E2559. [PMID: 32911852 PMCID: PMC7563767 DOI: 10.3390/cancers12092559] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 12/21/2022] Open
Abstract
Endometrial cancer (EC) remains one of the most common cancers of the female reproductive system. Epidemiological and clinical data implicate insulin resistance (IR) and its accompanying hyperinsulinemia as key factors in the development of EC. MicroRNAs (miRNAs) are short molecules of non-coding endogenous RNA that function as post-transcriptional regulators. Accumulating evidence has shown that the miRNA expression pattern is also likely to be associated with EC risk factors. The aim of this work was the verification of the relationships between IR, EC, and miRNA, and, as based on the literature data, elucidation of miRNA's potential utility for EC prevention in IR patients. The pathways affected in IR relate to the insulin receptors, insulin-like growth factors and their receptors, insulin-like growth factor binding proteins, sex hormone-binding globulin, and estrogens. Herein, we present and discuss arguments for miRNAs as a plausible molecular link between IR and EC development. Specifically, our careful literature search indicated that dysregulation of at least 13 miRNAs has been ascribed to both conditions. We conclude that there is a reasonable possibility for miRNAs to become a predictive factor of future EC in IR patients.
Collapse
Affiliation(s)
- Iwona Sidorkiewicz
- Clinical Research Centre, Medical University of Białystok, M. Skłodowskiej-Curie 24a, 15-276 Białystok, Poland; (M.N.); (A.K.)
| | - Maciej Jóźwik
- Department of Gynecology and Gynecologic Oncology, Medical University of Białystok, M. Skłodowskiej-Curie 24a, 15-276 Białystok, Poland;
| | - Magdalena Niemira
- Clinical Research Centre, Medical University of Białystok, M. Skłodowskiej-Curie 24a, 15-276 Białystok, Poland; (M.N.); (A.K.)
| | - Adam Krętowski
- Clinical Research Centre, Medical University of Białystok, M. Skłodowskiej-Curie 24a, 15-276 Białystok, Poland; (M.N.); (A.K.)
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Białystok, M. Skłodowskiej-Curie 24a, 15-276 Białystok, Poland
| |
Collapse
|
20
|
Higashi Y, Sukhanov S, Shai SY, Danchuk S, Snarski P, Li Z, Hou X, Hamblin MH, Woods TC, Wang M, Wang D, Yu H, Korthuis RJ, Yoshida T, Delafontaine P. Endothelial deficiency of insulin-like growth factor-1 receptor reduces endothelial barrier function and promotes atherosclerosis in Apoe-deficient mice. Am J Physiol Heart Circ Physiol 2020; 319:H730-H743. [PMID: 32795184 DOI: 10.1152/ajpheart.00064.2020] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Insulin-like growth factor-1 (IGF-1) decreases atherosclerosis in apolipoprotein E (Apoe)-deficient mice when administered systemically. However, mechanisms for its atheroprotective effect are not fully understood. We generated endothelium-specific IGF-1 receptor (IGF1R)-deficient mice on an Apoe-deficient background to assess effects of IGF-1 on the endothelium in the context of hyperlipidemia-induced atherosclerosis. Endothelial deficiency of IGF1R promoted atherosclerotic burden, when animals were fed on a high-fat diet for 12 wk or normal chow for 12 mo. Under the normal chow feeding condition, the vascular relaxation response to acetylcholine was increased in the endothelial IGF1R-deficient aorta; however, feeding of a high-fat diet substantially attenuated the relaxation response, and there was no difference between endothelial IGF1R-deficient and control mice. The endothelium and its intercellular junctions provide a barrier function to the vasculature. In human aortic endothelial cells, IGF-1 upregulated occludin, claudin 5, VE-cadherin, JAM-A, and CD31 expression levels, and vice versa, specific IGF1R inhibitor, picropodophyllin, an IGF1R-neutralizing antibody (αIR3), or siRNA to IGF1R abolished the IGF-1 effects on junction and adherens proteins, suggesting that IGF-1 promoted endothelial barrier function. Moreover, endothelial transwell permeability assays indicated that inhibition of IGF-1 signaling elevated solute permeability through the monolayer of human aortic endothelial cells. In summary, endothelial IGF1R deficiency increases atherosclerosis, and IGF-1 positively regulates tight junction protein and adherens junction protein levels and endothelial barrier function. Our findings suggest that the elevation of the endothelial junction protein level is, at least in part, the mechanism for antiatherogenic effects of IGF-1.NEW & NOTEWORTHY Endothelial insulin-like growth factor-1 (IGF-1) receptor deficiency significantly elevated atherosclerotic burden in apolipoprotein E-deficient mice, mediated at least in part by downregulation of intercellular junction proteins and, thus, elevated endothelial permeability. This study revealed a novel role for IGF-1 in supporting endothelial barrier function. These findings suggest that IGF-1's ability to promote endothelial barrier function may offer a novel therapeutic strategy for vascular diseases such as atherosclerosis.
Collapse
Affiliation(s)
- Yusuke Higashi
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Sergiy Sukhanov
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Shaw-Yung Shai
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Svitlana Danchuk
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Patricia Snarski
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Zhaohui Li
- Department of Radiology, University of Missouri School of Medicine, Columbia, Missouri
| | - Xuwei Hou
- Department of Medicine, University of Missouri School of Medicine, Columbia, Missouri
| | - Milton H Hamblin
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | - T Cooper Woods
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Meifang Wang
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - Derek Wang
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - Hong Yu
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - Ronald J Korthuis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - Tadashi Yoshida
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Patrice Delafontaine
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| |
Collapse
|
21
|
Steffensen LB, Conover CA, Oxvig C. PAPP-A and the IGF system in atherosclerosis: what's up, what's down? Am J Physiol Heart Circ Physiol 2019; 317:H1039-H1049. [PMID: 31518159 PMCID: PMC6879922 DOI: 10.1152/ajpheart.00395.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/23/2019] [Accepted: 08/23/2019] [Indexed: 12/26/2022]
Abstract
Pregnancy-associated plasma protein-A (PAPP-A) is a metalloproteinase with a well-established role in releasing bioactive insulin-like growth factor-1 (IGF-1) from IGF-binding protein-2, -4, and -5 by proteolytic processing of these. The IGF system has repeatedly been suggested to be involved in the pathology of atherosclerosis, and both PAPP-A and IGF-1 are proposed biomarkers and therapeutic targets for this disease. Several experimental approaches based on atherosclerosis mouse models have been undertaken to obtain causative and mechanistic insight to the role of these molecules in atherogenesis. However, reports seem conflicting. The literature suggests that PAPP-A is detrimental, while IGF-1 is beneficial. This raises important questions that need to be addressed. Here we summarize the various studies and discuss potential underlying explanations for this seemingly inconsistency with the objective of better understanding complexities and limitations when manipulating the IGF system in mouse models of atherosclerosis. A debate clarifying what's up and what's down is highly warranted going forward with the ultimate goal of improving atherosclerosis therapy by targeting the IGF system.
Collapse
Affiliation(s)
- Lasse B Steffensen
- Centre for Individualized Medicine in Arterial Diseases, Odense University Hospital, Odense, Denmark
| | | | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| |
Collapse
|
22
|
Abstract
Cardiovascular diseases are the most prominent maladies in aging societies. Indeed, aging promotes the structural and functional declines of both the heart and the blood circulation system. In this review, we revise the contribution of known longevity pathways to cardiovascular health and delineate the possibilities to interfere with them. In particular, we evaluate autophagy, the intracellular catabolic recycling system associated with life- and health-span extension. We present genetic models, pharmacological interventions, and dietary strategies that block, reduce, or enhance autophagy upon age-related cardiovascular deterioration. Caloric restriction or caloric restriction mimetics like metformin, spermidine, and rapamycin (all of which trigger autophagy) are among the most promising cardioprotective interventions during aging. We conclude that autophagy is a fundamental process to ensure cardiac and vascular health during aging and outline its putative therapeutic importance.
Collapse
Affiliation(s)
- Mahmoud Abdellatif
- From the Department of Cardiology, Medical University of Graz, Austria (M.A., S.S.)
| | - Simon Sedej
- From the Department of Cardiology, Medical University of Graz, Austria (M.A., S.S.).,BioTechMed Graz, Austria (S.S., D.C.-G., F.M.)
| | - Didac Carmona-Gutierrez
- BioTechMed Graz, Austria (S.S., D.C.-G., F.M.).,Institute of Molecular Biosciences, NAWI Graz, University of Graz, Austria (D.C.-G., F.M.)
| | - Frank Madeo
- BioTechMed Graz, Austria (S.S., D.C.-G., F.M.).,Institute of Molecular Biosciences, NAWI Graz, University of Graz, Austria (D.C.-G., F.M.)
| | - Guido Kroemer
- Equipe 11 Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France (G.K.).,Cell Biology and Metabolomics Platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France (G.K.).,INSERM, U1138, Paris, France (G.K.).,Université Paris Descartes, Sorbonne Paris Cité, France (G.K.).,Université Pierre et Marie Curie, Paris, France (G.K.).,Pôle de Biologie, Hôpital Européen Georges Pompidou, Paris, France (G.K.).,Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden (G.K.)
| |
Collapse
|
23
|
Homozygous receptors for insulin and not IGF-1 accelerate intimal hyperplasia in insulin resistance and diabetes. Nat Commun 2019; 10:4427. [PMID: 31562314 PMCID: PMC6765023 DOI: 10.1038/s41467-019-12368-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/03/2019] [Indexed: 11/08/2022] Open
Abstract
Insulin and IGF-1 actions in vascular smooth muscle cells (VSMC) are associated with accelerated arterial intima hyperplasia and restenosis after angioplasty, especially in diabetes. To distinguish their relative roles, we delete insulin receptor (SMIRKO) or IGF-1 receptor (SMIGF1RKO) in VSMC and in mice. Here we report that intima hyperplasia is attenuated in SMIRKO mice, but not in SMIGF1RKO mice. In VSMC, deleting IGF1R increases homodimers of IR, enhances insulin binding, stimulates p-Akt and proliferation, but deleting IR decreases responses to insulin and IGF-1. Studies using chimeras of IR(extracellular domain)/IGF1R(intracellular-domain) or IGF1R(extracellular domain)/IR(intracellular-domain) demonstrate homodimer IRα enhances insulin binding and signaling which is inhibited by IGF1Rα. RNA-seq identifies hyaluronan synthase2 as a target of homo-IR, with its expression increases by IR activation in SMIGF1RKO mice and decreases in SMIRKO mice. Enhanced intima hyperplasia in diabetes is mainly due to insulin signaling via homo-IR, associated with increased Has2 expression.
Collapse
|
24
|
Higashi Y, Gautam S, Delafontaine P, Sukhanov S. IGF-1 and cardiovascular disease. Growth Horm IGF Res 2019; 45:6-16. [PMID: 30735831 PMCID: PMC6504961 DOI: 10.1016/j.ghir.2019.01.002] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 10/17/2018] [Accepted: 01/30/2019] [Indexed: 12/14/2022]
Abstract
Atherosclerosis is an inflammatory arterial pathogenic condition, which leads to ischemic cardiovascular diseases, such as coronary artery disease and myocardial infarction, stroke, and peripheral arterial disease. Atherosclerosis is a multifactorial disorder and its pathophysiology is highly complex. Changes in expression of multiple genes coupled with environmental and lifestyle factors initiate cascades of adverse events involving multiple types of cells (e.g. vascular endothelial cells, smooth muscle cells, and macrophages). IGF-1 is a pleiotropic factor, which is found in the circulation (endocrine IGF-1) and is also produced locally in arteries (endothelial cells and smooth muscle cells). IGF-1 exerts a variety of effects on these cell types in the context of the pathogenesis of atherosclerosis. In fact, there is an increasing body of evidence suggesting that IGF-1 has beneficial effects on the biology of atherosclerosis. This review will discuss recent findings relating to clinical investigations on the relation between IGF-1 and cardiovascular disease and basic research using animal models of atherosclerosis that have elucidated some of the mechanisms underlying atheroprotective effects of IGF-1.
Collapse
Affiliation(s)
- Yusuke Higashi
- Department of Medicine, School of Medicine, University of Missouri, Columbia, MO, United States; Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States.
| | - Sandeep Gautam
- Department of Medicine, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Patrick Delafontaine
- Department of Medicine, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Sergiy Sukhanov
- Department of Medicine, School of Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
25
|
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: 1.5] [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
|
26
|
Haywood NJ, Slater TA, Matthews CJ, Wheatcroft SB. The insulin like growth factor and binding protein family: Novel therapeutic targets in obesity & diabetes. Mol Metab 2018; 19:86-96. [PMID: 30392760 PMCID: PMC6323188 DOI: 10.1016/j.molmet.2018.10.008] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/12/2018] [Accepted: 10/18/2018] [Indexed: 12/12/2022] Open
Abstract
Background Recent changes in nutrition and lifestyle have provoked an unprecedented increase in the prevalence of obesity and metabolic disorders. Recognition of the adverse effects on health has prompted intense efforts to understand the molecular determinants of insulin sensitivity and dysglycemia. In many respects, actions of insulin-like growth factors (IGFs) mirror those of insulin in metabolic regulation. Unlike insulin, however, the bioactivity of IGFs is regulated by a family of seven high-affinity binding proteins (IGFBPs) which confer temporospatial modulation with implications for metabolic homeostasis. In addition, evidence is accumulating that IGF-independent actions of certain of the IGFBPs can directly modulate insulin sensitivity. Scope of review In this review, we discuss the experimental data indicating a critical role for IGF/IGFBP axis in metabolic regulation. We highlight key discoveries through which IGFBPs have emerged as biomarkers or putative therapeutic targets in obesity and diabetes. Major conclusions Growing evidence suggests that several components of the IGF-IGFBP system could be explored for therapeutic potential in metabolic disorders. Both IGFBP-1 and IGFBP-2 have been favorably linked with insulin sensitivity in humans and preclinical data implicate direct involvement in the molecular regulation of insulin signaling and adiposity respectively. Further studies are warranted to evaluate clinical translation of these findings.
Collapse
Affiliation(s)
- Natalie J Haywood
- Division of Cardiovascular and Diabetes Research, Leeds Multidisciplinary Cardiovascular Research Centre, Faculty of Medicine and Health, University of Leeds, United Kingdom
| | - Thomas A Slater
- Division of Cardiovascular and Diabetes Research, Leeds Multidisciplinary Cardiovascular Research Centre, Faculty of Medicine and Health, University of Leeds, United Kingdom
| | - Connor J Matthews
- Division of Cardiovascular and Diabetes Research, Leeds Multidisciplinary Cardiovascular Research Centre, Faculty of Medicine and Health, University of Leeds, United Kingdom
| | - Stephen B Wheatcroft
- Division of Cardiovascular and Diabetes Research, Leeds Multidisciplinary Cardiovascular Research Centre, Faculty of Medicine and Health, University of Leeds, United Kingdom.
| |
Collapse
|
27
|
Hu J, Liu X, Chi J, Che K, Feng Y, Zhao S, Wang Z, Wang Y. Expressions of IGF-1, ERK, GLUT4, IRS-1 in metabolic syndrome complicated with colorectal cancer and their associations with the clinical characteristics of CRC. Cancer Biomark 2018; 21:883-891. [PMID: 29504525 DOI: 10.3233/cbm-170942] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Epidemiological data have revealed that colorectal cancer (CRC) risk is increased in patients with Metabolic syndrome. OBJECTIVE To explore the expressions of IGF-1, ERK, GLUT4, IRS-1 in MS patients with CRC and their associations with the clinical characteristics of CRC. METHODS We investigated the expressions of IGF-1, ERK, GLUT4 and IRS-1 in greater omental adipose tissues of 168 MS patients with/without CRC, 85 CRC patients without MS and 98 healthy controls by RT-PCR, and analyzed the relationships between their expressions and clinical characteristics of CRC. RESULTS The expression levels of IGF-1 and ERK in MS patients with/without CRC were higher while the expression levels of GLUT4 were lower compared with CRC patients without MS and healthy controls (P< 0.01). The expression levels of IGF-1 and ERK in MS patients with CRC were higher while expression levels of GLUT4 were lower compared to MS patients without CRC (P< 0.01). Expression levels of ERK, IGF-1, GLUT4 were associated with clinical characteristics of CRC, including tumor size, distant metastasis and advanced stages (III/IV) (P< 0.05). CONCLUSIONS Expressions of IGF-1, ERK and GLUT4 in greater omental adipose tissues might be useful biomarkers and predictive targets in the diagnosis of CRC.
Collapse
Affiliation(s)
- Jianxia Hu
- The Laboratory of Thyroid Disease, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,The Laboratory of Thyroid Disease, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaoyi Liu
- The Center of Breast Disease, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,The Laboratory of Thyroid Disease, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jingwei Chi
- The Laboratory of Thyroid Disease, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Kui Che
- The Laboratory of Thyroid Disease, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yan Feng
- Endocrinology Department, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shihua Zhao
- The Laboratory of Thyroid Disease, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zhongchao Wang
- Endocrinology Department, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yangang Wang
- Endocrinology Department, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
28
|
Sengupta A, Patel PA, Yuldasheva NY, Mughal RS, Galloway S, Viswambharan H, Walker AMN, Aziz A, Smith J, Ali N, Mercer BN, Imrie H, Sukumar P, Wheatcroft SB, Kearney MT, Cubbon RM. Endothelial Insulin Receptor Restoration Rescues Vascular Function in Male Insulin Receptor Haploinsufficient Mice. Endocrinology 2018; 159:2917-2925. [PMID: 29796592 PMCID: PMC6047419 DOI: 10.1210/en.2018-00215] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/25/2018] [Indexed: 11/19/2022]
Abstract
Reduced systemic insulin signaling promotes endothelial dysfunction and diminished endogenous vascular repair. We investigated whether restoration of endothelial insulin receptor expression could rescue this phenotype. Insulin receptor knockout (IRKO) mice were crossed with mice expressing a human insulin receptor endothelial cell-specific overexpression (hIRECO) to produce IRKO-hIRECO progeny. No metabolic differences were noted between IRKO and IRKO-hIRECO mice in glucose and insulin tolerance tests. In contrast with control IRKO littermates, IRKO-hIRECO mice exhibited normal blood pressure and aortic vasodilatation in response to acetylcholine, comparable to parameters noted in wild type littermates. These phenotypic changes were associated with increased basal- and insulin-stimulated nitric oxide production. IRKO-hIRECO mice also demonstrated normalized endothelial repair after denuding arterial injury, which was associated with rescued endothelial cell migration in vitro but not with changes in circulating progenitor populations or culture-derived myeloid angiogenic cells. These data show that restoration of endothelial insulin receptor expression alone is sufficient to prevent the vascular dysfunction caused by systemically reduced insulin signaling.
Collapse
Affiliation(s)
- Anshuman Sengupta
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Leeds, United Kingdom
| | - Peysh A Patel
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Leeds, United Kingdom
| | - Nadira Y Yuldasheva
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Leeds, United Kingdom
| | - Romana S Mughal
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Leeds, United Kingdom
| | - Stacey Galloway
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Leeds, United Kingdom
| | - Hema Viswambharan
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Leeds, United Kingdom
| | - Andrew M N Walker
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Leeds, United Kingdom
| | - Amir Aziz
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Leeds, United Kingdom
| | - Jessica Smith
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Leeds, United Kingdom
| | - Noman Ali
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Leeds, United Kingdom
| | - Ben N Mercer
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Leeds, United Kingdom
| | - Helen Imrie
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Leeds, United Kingdom
| | - Piruthivi Sukumar
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Leeds, United Kingdom
| | - Stephen B Wheatcroft
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Leeds, United Kingdom
| | - Mark T Kearney
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Leeds, United Kingdom
| | - Richard M Cubbon
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Leeds, United Kingdom
- Correspondence: Richard M. Cubbon, MBChB, PhD, Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom. E-mail:
| |
Collapse
|
29
|
Carr CA, Dodd MS, Heather LC. Bernard and Joan Marshall Awards at the autumn meeting of the British Society for Cardiovascular Research 2017. Heart 2018; 104:1383-1384. [PMID: 29925542 DOI: 10.1136/heartjnl-2018-313271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Affiliation(s)
- Carolyn A Carr
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Michael S Dodd
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Lisa C Heather
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
30
|
Kanter JE, Kramer F, Barnhart S, Duggan JM, Shimizu-Albergine M, Kothari V, Chait A, Bouman SD, Hamerman JA, Hansen BF, Olsen GS, Bornfeldt KE. A Novel Strategy to Prevent Advanced Atherosclerosis and Lower Blood Glucose in a Mouse Model of Metabolic Syndrome. Diabetes 2018; 67:946-959. [PMID: 29483182 PMCID: PMC5909997 DOI: 10.2337/db17-0744] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Accepted: 02/01/2018] [Indexed: 12/20/2022]
Abstract
Cardiovascular disease caused by atherosclerosis is the leading cause of mortality associated with type 2 diabetes and metabolic syndrome. Insulin therapy is often needed to improve glycemic control, but it does not clearly prevent atherosclerosis. Upon binding to the insulin receptor (IR), insulin activates distinct arms of downstream signaling. The IR-Akt arm is associated with blood glucose lowering and beneficial effects, whereas the IR-Erk arm might exert less desirable effects. We investigated whether selective activation of the IR-Akt arm, leaving the IR-Erk arm largely inactive, would result in protection from atherosclerosis in a mouse model of metabolic syndrome. The insulin mimetic peptide S597 lowered blood glucose and activated Akt in insulin target tissues, mimicking insulin's effects, but only weakly activated Erk and even prevented insulin-induced Erk activation. Strikingly, S597 retarded atherosclerotic lesion progression through a process associated with protection from leukocytosis, thereby reducing lesional accumulation of inflammatory Ly6Chi monocytes. S597-mediated protection from leukocytosis was accompanied by reduced numbers of the earliest bone marrow hematopoietic stem cells and reduced IR-Erk activity in hematopoietic stem cells. This study provides a conceptually novel treatment strategy for advanced atherosclerosis associated with metabolic syndrome and type 2 diabetes.
Collapse
Affiliation(s)
- Jenny E Kanter
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Farah Kramer
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Shelley Barnhart
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Jeffrey M Duggan
- Department of Immunology, University of Washington, Seattle, WA
- Benaroya Research Institute, Seattle, WA
| | - Masami Shimizu-Albergine
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Vishal Kothari
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Alan Chait
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | | | - Jessica A Hamerman
- Department of Immunology, University of Washington, Seattle, WA
- Benaroya Research Institute, Seattle, WA
| | - Bo F Hansen
- Insulin Biology Department, Novo Nordisk A/S, Måløv, Denmark
| | - Grith S Olsen
- Insulin Biology Department, Novo Nordisk A/S, Måløv, Denmark
| | - Karin E Bornfeldt
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
- Department of Pathology, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| |
Collapse
|
31
|
Guan SP, Lam ATL, Newman JP, Chua KLM, Kok CYL, Chong ST, Chua MLK, Lam PYP. Matrix metalloproteinase-1 facilitates MSC migration via cleavage of IGF-2/IGFBP2 complex. FEBS Open Bio 2017; 8:15-26. [PMID: 29321953 PMCID: PMC5757182 DOI: 10.1002/2211-5463.12330] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/26/2017] [Accepted: 09/28/2017] [Indexed: 12/16/2022] Open
Abstract
The specific mechanism underlying the tumor tropism of human mesenchymal stem cells (MSCs) for cancer is not well defined. We previously showed that the migration potential of MSCs correlated with the expression and protease activity of matrix metalloproteinase (MMP)‐1. Furthermore, highly tumor‐tropic MSCs expressed higher levels of MMP‐1 and insulin‐like growth factor (IGF)‐2 than poorly migrating MSCs. In this study, we examined the functional roles of IGF‐2 and MMP‐1 in mediating the tumor tropism of MSCs. Exogenous addition of either recombinant IGF‐2 or MMP‐1 could stimulate MSC migration. The correlation between IGF‐2, MMP‐1 expression, and MSC migration suggests that MMP‐1 may play a role in regulating MSC migration via the IGF‐2 signaling cascade. High concentrations of IGF binding proteins (IGFBPs) can inhibit IGF‐stimulated functions by blocking its binding to its receptors and proteolysis of IGFBP is an important mechanism for the regulation of IGF signaling. We thus hypothesized that MMP‐1 acts as an IGFBP2 proteinase, resulting in the cleavage of IGF‐2/IGFBP2 complex and extracellular release of free IGF‐2. Indeed, our results showed that conditioned media from highly migrating MSCs, which expressed high levels of MMP‐1, cleaved the IGF‐2/IGFBP2 complex. Taken together, these results showed that the MMP‐1 secreted by highly tumor‐tropic MSCs cleaved IGF‐2/IGFBP2 complex. Free IGF‐2 released from the complex may facilitate MSC migration toward tumor.
Collapse
Affiliation(s)
- Shou P Guan
- Laboratory of Cancer Gene Therapy, Cellular and Molecular Research Division Humphrey Oei Institute of Cancer Research National Cancer Center Singapore Singapore
| | - Alan T L Lam
- Laboratory of Cancer Gene Therapy, Cellular and Molecular Research Division Humphrey Oei Institute of Cancer Research National Cancer Center Singapore Singapore.,Present address: BTIASTAR Centros Singapore
| | - Jennifer P Newman
- Laboratory of Cancer Gene Therapy, Cellular and Molecular Research Division Humphrey Oei Institute of Cancer Research National Cancer Center Singapore Singapore.,Present address: Lonza Biologics Tuas Pte Ltd Singapore
| | - Kevin L M Chua
- Division of Radiation Oncology National Cancer Center Singapore Singapore
| | - Catherine Y L Kok
- Laboratory of Cancer Gene Therapy, Cellular and Molecular Research Division Humphrey Oei Institute of Cancer Research National Cancer Center Singapore Singapore
| | - Siao T Chong
- Laboratory of Cancer Gene Therapy, Cellular and Molecular Research Division Humphrey Oei Institute of Cancer Research National Cancer Center Singapore Singapore
| | - Melvin L K Chua
- Division of Radiation Oncology National Cancer Center Singapore Singapore.,Oncology Academic Program Duke-NUS Graduate Medical School Singapore Singapore
| | - Paula Y P Lam
- Laboratory of Cancer Gene Therapy, Cellular and Molecular Research Division Humphrey Oei Institute of Cancer Research National Cancer Center Singapore Singapore.,Cancer and Stem Cells Biology Program Duke-NUS Graduate Medical School Singapore Singapore.,Department of Physiology Yong Loo Lin School of Medicine National University of Singapore Singapore
| |
Collapse
|
32
|
Watt NT, Gage MC, Patel PA, Viswambharan H, Sukumar P, Galloway S, Yuldasheva NY, Imrie H, Walker AMN, Griffin KJ, Makava N, Skromna A, Bridge K, Beech DJ, Schurmans S, Wheatcroft SB, Kearney MT, Cubbon RM. Endothelial SHIP2 Suppresses Nox2 NADPH Oxidase-Dependent Vascular Oxidative Stress, Endothelial Dysfunction, and Systemic Insulin Resistance. Diabetes 2017; 66:2808-2821. [PMID: 28830894 DOI: 10.2337/db17-0062] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 08/04/2017] [Indexed: 11/13/2022]
Abstract
Shc homology 2-containing inositol 5' phosphatase-2 (SHIP2) is a lipid phosphatase that inhibits insulin signaling downstream of phosphatidylinositol 3-kinase (PI3K); its role in vascular function is poorly understood. To examine its role in endothelial cell (EC) biology, we generated mice with catalytic inactivation of one SHIP2 allele selectively in ECs (ECSHIP2Δ/+). Hyperinsulinemic-euglycemic clamping studies revealed that ECSHIP2Δ/+ was resistant to insulin-stimulated glucose uptake in adipose tissue and skeletal muscle compared with littermate controls. ECs from ECSHIP2Δ/+ mice had increased basal expression and activation of PI3K downstream targets, including Akt and endothelial nitric oxide synthase, although incremental activation by insulin and shear stress was impaired. Insulin-mediated vasodilation was blunted in ECSHIP2Δ/+ mice, as was aortic nitric oxide bioavailability. Acetylcholine-induced vasodilation was also impaired in ECSHIP2Δ/+ mice, which was exaggerated in the presence of a superoxide dismutase/catalase mimetic. Superoxide abundance was elevated in ECSHIP2Δ/+ ECs and was suppressed by PI3K and NADPH oxidase 2 inhibitors. These findings were phenocopied in healthy human ECs after SHIP2 silencing. Our data suggest that endothelial SHIP2 is required to maintain normal systemic glucose homeostasis and prevent oxidative stress-induced endothelial dysfunction.
Collapse
Affiliation(s)
- Nicole T Watt
- Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, U.K
| | - Matthew C Gage
- Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, U.K
| | - Peysh A Patel
- Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, U.K
| | - Hema Viswambharan
- Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, U.K
| | - Piruthivi Sukumar
- Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, U.K
| | - Stacey Galloway
- Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, U.K
| | - Nadira Y Yuldasheva
- Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, U.K
| | - Helen Imrie
- Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, U.K
| | - Andrew M N Walker
- Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, U.K
| | - Kathryn J Griffin
- Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, U.K
| | - Natalia Makava
- Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, U.K
| | - Anna Skromna
- Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, U.K
| | - Katherine Bridge
- Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, U.K
| | - David J Beech
- Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, U.K
| | - Stéphane Schurmans
- Laboratory of Functional Genetics, GIGA Research Centre, Université de Liège, Liège, Belgium
| | - Stephen B Wheatcroft
- Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, U.K
| | - Mark T Kearney
- Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, U.K.
| | - Richard M Cubbon
- Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, U.K
| |
Collapse
|
33
|
Belfiore A, Malaguarnera R, Vella V, Lawrence MC, Sciacca L, Frasca F, Morrione A, Vigneri R. Insulin Receptor Isoforms in Physiology and Disease: An Updated View. Endocr Rev 2017; 38:379-431. [PMID: 28973479 PMCID: PMC5629070 DOI: 10.1210/er.2017-00073] [Citation(s) in RCA: 266] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/13/2017] [Indexed: 02/08/2023]
Abstract
The insulin receptor (IR) gene undergoes differential splicing that generates two IR isoforms, IR-A and IR-B. The physiological roles of IR isoforms are incompletely understood and appear to be determined by their different binding affinities for insulin-like growth factors (IGFs), particularly for IGF-2. Predominant roles of IR-A in prenatal growth and development and of IR-B in metabolic regulation are well established. However, emerging evidence indicates that the differential expression of IR isoforms may also help explain the diversification of insulin and IGF signaling and actions in various organs and tissues by involving not only different ligand-binding affinities but also different membrane partitioning and trafficking and possibly different abilities to interact with a variety of molecular partners. Of note, dysregulation of the IR-A/IR-B ratio is associated with insulin resistance, aging, and increased proliferative activity of normal and neoplastic tissues and appears to sustain detrimental effects. This review discusses novel information that has generated remarkable progress in our understanding of the physiology of IR isoforms and their role in disease. We also focus on novel IR ligands and modulators that should now be considered as an important strategy for better and safer treatment of diabetes and cancer and possibly other IR-related diseases.
Collapse
Affiliation(s)
- Antonino Belfiore
- Endocrinology, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Roberta Malaguarnera
- Endocrinology, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Veronica Vella
- School of Human and Social Sciences, University Kore of Enna, via della Cooperazione, 94100 Enna, Italy
| | - Michael C. Lawrence
- Structural Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Laura Sciacca
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy
| | - Francesco Frasca
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy
| | - Andrea Morrione
- Department of Urology and Biology of Prostate Cancer Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Riccardo Vigneri
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy
| |
Collapse
|
34
|
Viswambharan H, Yuldasheva NY, Sengupta A, Imrie H, Gage MC, Haywood N, Walker AM, Skromna A, Makova N, Galloway S, Shah P, Sukumar P, Porter KE, Grant PJ, Shah AM, Santos CX, Li J, Beech DJ, Wheatcroft SB, Cubbon RM, Kearney MT. Selective Enhancement of Insulin Sensitivity in the Endothelium In Vivo Reveals a Novel Proatherosclerotic Signaling Loop. Circ Res 2017; 120:784-798. [DOI: 10.1161/circresaha.116.309678] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 11/30/2016] [Accepted: 12/05/2016] [Indexed: 12/21/2022]
Abstract
Rationale:
In the endothelium, insulin stimulates endothelial NO synthase (eNOS) to generate the antiatherosclerotic signaling radical NO. Insulin-resistant type 2 diabetes mellitus is associated with reduced NO availability and accelerated atherosclerosis. The effect of enhancing endothelial insulin sensitivity on NO availability is unclear.
Objective:
To answer this question, we generated a mouse with endothelial cell (EC)–specific overexpression of the human insulin receptor (hIRECO) using the Tie2 promoter–enhancer.
Methods and Results:
hIRECO demonstrated significant endothelial dysfunction measured by blunted endothelium-dependent vasorelaxation to acetylcholine, which was normalized by a specific Nox2 NADPH oxidase inhibitor. Insulin-stimulated phosphorylation of protein kinase B was increased in hIRECO EC as was Nox2 NADPH oxidase–dependent generation of superoxide, whereas insulin-stimulated and shear stress–stimulated eNOS activations were blunted. Phosphorylation at the inhibitory residue Y657 of eNOS and expression of proline-rich tyrosine kinase 2 that phosphorylates this residue were significantly higher in hIRECO EC. Inhibition of proline-rich tyrosine kinase 2 improved insulin-induced and shear stress–induced eNOS activation in hIRECO EC.
Conclusions:
Enhancing insulin sensitivity specifically in EC leads to a paradoxical decline in endothelial function, mediated by increased tyrosine phosphorylation of eNOS and excess Nox2-derived superoxide. Increased EC insulin sensitivity leads to a proatherosclerotic imbalance between NO and superoxide. Inhibition of proline-rich tyrosine kinase 2 restores insulin-induced and shear stress–induced NO production. This study demonstrates for the first time that increased endothelial insulin sensitivity leads to a proatherosclerotic imbalance between NO and superoxide.
Collapse
Affiliation(s)
- Hema Viswambharan
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Nadira Y. Yuldasheva
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Anshuman Sengupta
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Helen Imrie
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Matthew C. Gage
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Natalie Haywood
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Andrew M.N. Walker
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Anna Skromna
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Natallia Makova
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Stacey Galloway
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Pooja Shah
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Piruthivi Sukumar
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Karen E. Porter
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Peter J. Grant
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Ajay M. Shah
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Celio X.C. Santos
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Jing Li
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - David J. Beech
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Stephen B. Wheatcroft
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Richard M. Cubbon
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Mark T. Kearney
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| |
Collapse
|
35
|
López IP, Piñeiro-Hermida S, Pais RS, Torrens R, Hoeflich A, Pichel JG. Involvement of Igf1r in Bronchiolar Epithelial Regeneration: Role during Repair Kinetics after Selective Club Cell Ablation. PLoS One 2016; 11:e0166388. [PMID: 27861515 PMCID: PMC5115747 DOI: 10.1371/journal.pone.0166388] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 10/27/2016] [Indexed: 12/14/2022] Open
Abstract
Regeneration of lung epithelium is vital for maintaining airway function and integrity. An imbalance between epithelial damage and repair is at the basis of numerous chronic lung diseases such as asthma, COPD, pulmonary fibrosis and lung cancer. IGF (Insulin-like Growth Factors) signaling has been associated with most of these respiratory pathologies, although their mechanisms of action in this tissue remain poorly understood. Expression profiles analyses of IGF system genes performed in mouse lung support their functional implication in pulmonary ontogeny. Immuno-localization revealed high expression levels of Igf1r (Insulin-like Growth Factor 1 Receptor) in lung epithelial cells, alveolar macrophages and smooth muscle. To further understand the role of Igf1r in pulmonary homeostasis, two distinct lung epithelial-specific Igf1r mutant mice were generated and studied. The lack of Igf1r disturbed airway epithelial differentiation in adult mice, and revealed enhanced proliferation and altered morphology in distal airway club cells. During recovery after naphthalene-induced club cell injury, the kinetics of terminal bronchiolar epithelium regeneration was hindered in Igf1r mutants, revealing increased proliferation and delayed differentiation of club and ciliated cells. Amid airway restoration, lungs of Igf1r deficient mice showed increased levels of Igf1, Insr, Igfbp3 and epithelial precursor markers, reduced amounts of Scgb1a1 protein, and alterations in IGF signaling mediators. These results support the role of Igf1r in controlling the kinetics of cell proliferation and differentiation during pulmonary airway epithelial regeneration after injury.
Collapse
Affiliation(s)
- Icíar P López
- Centro de Investigación Biomédica de la Rioja (CIBIR), Fundación Rioja Salud, Logroño, Spain
| | - Sergio Piñeiro-Hermida
- Centro de Investigación Biomédica de la Rioja (CIBIR), Fundación Rioja Salud, Logroño, Spain
| | - Rosete S Pais
- Centro de Investigación Biomédica de la Rioja (CIBIR), Fundación Rioja Salud, Logroño, Spain
| | - Raquel Torrens
- Centro de Investigación Biomédica de la Rioja (CIBIR), Fundación Rioja Salud, Logroño, Spain
| | - Andreas Hoeflich
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - José G Pichel
- Centro de Investigación Biomédica de la Rioja (CIBIR), Fundación Rioja Salud, Logroño, Spain
| |
Collapse
|
36
|
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: 2.9] [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
|
37
|
Milman S, Huffman DM, Barzilai N. The Somatotropic Axis in Human Aging: Framework for the Current State of Knowledge and Future Research. Cell Metab 2016; 23:980-989. [PMID: 27304500 PMCID: PMC4919980 DOI: 10.1016/j.cmet.2016.05.014] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 05/24/2016] [Accepted: 05/26/2016] [Indexed: 12/19/2022]
Abstract
Mutations resulting in reduced signaling of the growth hormone/insulin-like growth factor-1 (GH/IGF-1) axis are associated with increased life- and healthspan across model organisms. Similar findings have been noted in human cohorts with functional mutations in the somatotropic axis, suggesting that this pathway may also be relevant to human aging and protection from age-related diseases. While epidemiological data indicate that low circulating IGF-1 level may protect aging populations from cancer, results remain inconclusive regarding most other diseases. We propose that studies in humans and animals need to consider differences in sex, pathway function, organs, and time-specific effects of GH/IGF-1 signaling in order to better define the role of the somatotropic axis in aging. Agents that modulate signaling of the GH/IGF-1 pathway are available for human use, but before they can be implemented in clinical studies that target aging and age-related diseases, researchers need to address the challenges discussed in this Review.
Collapse
Affiliation(s)
- Sofiya Milman
- Department of Medicine, Division of Endocrinology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Medicine, Division of Geriatrics, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Derek M Huffman
- Department of Medicine, Division of Endocrinology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Nir Barzilai
- Department of Medicine, Division of Endocrinology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Medicine, Division of Geriatrics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
38
|
Pek SLT, Sum CF, Lin MX, Cheng AKS, Wong MTK, Lim SC, Tavintharan S. Circulating and visceral adipose miR-100 is down-regulated in patients with obesity and Type 2 diabetes. Mol Cell Endocrinol 2016; 427:112-23. [PMID: 26973292 DOI: 10.1016/j.mce.2016.03.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/04/2016] [Accepted: 03/08/2016] [Indexed: 11/30/2022]
Abstract
Obesity is a major public health problem conferring substantial excess risk for Type 2 diabetes (T2D). The role of microRNAs (miRNAs) in obesity and adipose tissue is not clearly defined. We hypothesize that circulating miRNA expression profiles vary according to differences in body mass index (BMI) and T2D and circulating miRNAs may reflect adipose tissue expression. Compared to healthy, lean individuals, circulating miR-100 was significantly lower in obese normoglycemic subjects and subjects with T2D. In visceral adipose tissue, expression of miR-100 was lower from obese subjects with T2D compared to obese subjects without T2D. miR-100 expression was significantly lower after adipogenic induction in human visceral, subcutaneous adipocytes and 3T3-L1 adipocytes. miR-100 reduced expression of mammalian target of rapamycin (mTOR) and Insulin Growth Factor Receptor (IGFR) directly. Differentiation of 3T3-L1 was accelerated by inhibition of miR-100 and reduced by miR-100 mimic transfection. Our data provide the first evidence of an association of circulating miR-100 with obesity and diabetes. Additionally, our in-vitro findings, and the miR-100 expression patterns in site-specific adipose tissue suggest miR-100 to modulate IGFR, mTOR and mediate adipogenesis.
Collapse
Affiliation(s)
| | - Chee Fang Sum
- Diabetes Centre, Khoo Teck Puat Hospital, 768828, Singapore; Division of Endocrinology, Khoo Teck Puat Hospital, 768828, Singapore.
| | | | | | | | - Su Chi Lim
- Clinical Research Unit, Khoo Teck Puat Hospital, 768828, Singapore; Diabetes Centre, Khoo Teck Puat Hospital, 768828, Singapore; Division of Endocrinology, Khoo Teck Puat Hospital, 768828, Singapore.
| | - Subramaniam Tavintharan
- Clinical Research Unit, Khoo Teck Puat Hospital, 768828, Singapore; Diabetes Centre, Khoo Teck Puat Hospital, 768828, Singapore; Division of Endocrinology, Khoo Teck Puat Hospital, 768828, Singapore.
| |
Collapse
|
39
|
Higashi Y, Sukhanov S, Shai SY, Danchuk S, Tang R, Snarski P, Li Z, Lobelle-Rich P, Wang M, Wang D, Yu H, Korthuis R, Delafontaine P. Insulin-Like Growth Factor-1 Receptor Deficiency in Macrophages Accelerates Atherosclerosis and Induces an Unstable Plaque Phenotype in Apolipoprotein E-Deficient Mice. Circulation 2016; 133:2263-78. [PMID: 27154724 DOI: 10.1161/circulationaha.116.021805] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 04/27/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND We have previously shown that systemic infusion of insulin-like growth factor-1 (IGF-1) exerts anti-inflammatory and antioxidant effects and reduces atherosclerotic burden in apolipoprotein E (Apoe)-deficient mice. Monocytes/macrophages express high levels of IGF-1 receptor (IGF1R) and play a pivotal role in atherogenesis, but the potential effects of IGF-1 on their function are unknown. METHODS AND RESULTS To determine mechanisms whereby IGF-1 reduces atherosclerosis and to explore the potential involvement of monocytes/macrophages, we created monocyte/macrophage-specific IGF1R knockout (MΦ-IGF1R-KO) mice on an Apoe(-/-) background. We assessed atherosclerotic burden, plaque features of stability, and monocyte recruitment to atherosclerotic lesions. Phenotypic changes of IGF1R-deficient macrophages were investigated in culture. MΦ-IGF1R-KO significantly increased atherosclerotic lesion formation, as assessed by Oil Red O staining of en face aortas and aortic root cross-sections, and changed plaque composition to a less stable phenotype, characterized by increased macrophage and decreased α-smooth muscle actin-positive cell population, fibrous cap thinning, and decreased collagen content. Brachiocephalic artery lesions of MΦ-IGF1R-KO mice had histological features implying plaque vulnerability. Macrophages isolated from MΦ-IGF1R-KO mice showed enhanced proinflammatory responses on stimulation by interferon-γ and oxidized low-density lipoprotein and elevated antioxidant gene expression levels. Moreover, IGF1R-deficient macrophages had decreased expression of ABCA1 and ABCG1 and reduced lipid efflux. CONCLUSIONS Our data indicate that macrophage IGF1R signaling suppresses macrophage and foam cell accumulation in lesions and reduces plaque vulnerability, providing a novel mechanism whereby IGF-1 exerts antiatherogenic effects.
Collapse
Affiliation(s)
- Yusuke Higashi
- From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.).
| | - Sergiy Sukhanov
- From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.)
| | - Shaw-Yung Shai
- From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.)
| | - Svitlana Danchuk
- From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.)
| | - Richard Tang
- From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.)
| | - Patricia Snarski
- From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.)
| | - Zhaohui Li
- From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.)
| | - Patricia Lobelle-Rich
- From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.)
| | - Meifang Wang
- From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.)
| | - Derek Wang
- From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.)
| | - Hong Yu
- From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.)
| | - Ronald Korthuis
- From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.)
| | - Patrice Delafontaine
- From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.)
| |
Collapse
|
40
|
Affiliation(s)
- Hong Lu
- From the Saha Cardiovascular Research Center, University of Kentucky, Lexington.
| | - Alan Daugherty
- From the Saha Cardiovascular Research Center, University of Kentucky, Lexington
| |
Collapse
|
41
|
Cubbon RM, Kearney MT, Wheatcroft SB. Endothelial IGF-1 Receptor Signalling in Diabetes and Insulin Resistance. Trends Endocrinol Metab 2016; 27:96-104. [PMID: 26712712 DOI: 10.1016/j.tem.2015.11.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 11/22/2015] [Accepted: 11/24/2015] [Indexed: 01/19/2023]
Abstract
Despite contemporary medical therapy, people with diabetes and insulin resistance experience substantially increased risk of cardiovascular events caused by atherosclerosis. Dysfunction of the endothelium is a key phase in early atherogenesis and represents a promising therapeutic target. The evolutionarily related insulin and insulin-like growth factor-1 (IGF-1) axes are implicated in the development of vascular disease. In this review, we summarise recent developments in our understanding of how modulating the IGF-1 axis influences vascular disease in the setting of insulin resistance. By contrasting data from models of altered insulin and/or IGF-1 signalling, we emphasise the complex spatiotemporal interplay of these systems in health and disease. We then discuss therapeutic opportunities, before detailing important gaps in our knowledge relevant to therapeutic translation.
Collapse
Affiliation(s)
- Richard M Cubbon
- Leeds Institute for Cardiovascular and Metabolic Medicine, LIGHT laboratories, University of Leeds, Clarendon Way, Leeds, LS2 9JT, UK
| | - Mark T Kearney
- Leeds Institute for Cardiovascular and Metabolic Medicine, LIGHT laboratories, University of Leeds, Clarendon Way, Leeds, LS2 9JT, UK.
| | - Stephen B Wheatcroft
- Leeds Institute for Cardiovascular and Metabolic Medicine, LIGHT laboratories, University of Leeds, Clarendon Way, Leeds, LS2 9JT, UK
| |
Collapse
|
42
|
Martins IJ. Anti-Aging Genes Improve Appetite Regulation and Reverse Cell Senescence and Apoptosis in Global Populations. ACTA ACUST UNITED AC 2016. [DOI: 10.4236/aar.2016.51002] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
43
|
Novel players in cardioprotection: Insulin like growth factor-1, angiotensin-(1–7) and angiotensin-(1–9). Pharmacol Res 2015; 101:41-55. [DOI: 10.1016/j.phrs.2015.06.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 06/27/2015] [Accepted: 06/28/2015] [Indexed: 12/14/2022]
|
44
|
Lin YY, Lee SD, Su CT, Cheng TL, Yang AL. Long-term treadmill training ameliorates endothelium-dependent vasorelaxation mediated by insulin and insulin-like growth factor-1 in hypertension. J Appl Physiol (1985) 2015; 119:663-9. [PMID: 26183483 DOI: 10.1152/japplphysiol.01062.2014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 07/10/2015] [Indexed: 11/22/2022] Open
Abstract
Dysfunction of insulin and insulin-like growth factor-1 (IGF-1) is associated with the pathophysiology of hypertension. The influence of long-term exercise on vascular dysfunction caused by hypertension remains unclear. We investigated whether long-term treadmill training improved insulin- and IGF-1-mediated vasorelaxation in hypertensive rats. Eight-week-old male spontaneously hypertensive rats (SHR) were randomly divided into sedentary and exercise (SHR-EX) groups. The SHR-EX group was trained on a treadmill for 60 min/day, 5 days/wk, for 8 wk. Wistar-Kyoto rats (WKY) were used as the normal control group. After training, aortic insulin- and IGF-1-mediated vasorelaxation was evaluated in organ baths. Additionally, the roles of phosphatidylinositol 3-kinase (PI3K), nitric oxide synthase (NOS), and aortic protein expression were examined in the three groups. Compared with sedentary SHR and WKY groups, insulin- and IGF-1-mediated vasorelaxation was significantly enhanced to a nearly normal level in the SHR-EX group. After endothelial denudation, blunted and comparable vasorelaxation was found among the three groups. Pretreatment with selective PI3K and NOS inhibitors attenuated insulin- and IGF-1-mediated vasorelaxation, and no significant difference was found among the three groups after the pretreatment. The aortic protein levels of the insulin receptor (IR), IGF-1 receptor (IGF-1R), insulin receptor substrate-1 (IRS-1), and endothelial NOS (eNOS) were also significantly increased in the SHR-EX group compared with the other two groups. These results suggested that treadmill training elicited the amelioration of endothelium-dependent insulin/IGF-1-mediated vasorelaxation partly via the increased activation of PI3K and NOS, as well as the enhancement of protein levels of IR, IGF-1R, IRS-1, and eNOS, in hypertension.
Collapse
Affiliation(s)
- Yi-Yuan Lin
- Graduate Institute of Clinical Medical Science and
| | - Shin-Da Lee
- Graduate Institute of Clinical Medical Science and Department of Physical Therapy, Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan; Department of Healthcare Administration, Asia University, Taichung, Taiwan
| | - Chia-Ting Su
- Department of Occupational Therapy, College of Medicine, Fu Jen Catholic University, Taipei, Taiwan
| | - Tsung-Lin Cheng
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; and
| | - Ai-Lun Yang
- Graduate Institute of Exercise Science, University of Taipei, Taipei, Taiwan
| |
Collapse
|
45
|
Sauvet F, Drogou C, Bougard C, Arnal PJ, Dispersyn G, Bourrilhon C, Rabat A, Van Beers P, Gomez-Merino D, Faraut B, Leger D, Chennaoui M. Vascular response to 1 week of sleep restriction in healthy subjects. A metabolic response? Int J Cardiol 2015; 190:246-55. [PMID: 25932797 DOI: 10.1016/j.ijcard.2015.04.119] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 03/02/2015] [Accepted: 04/15/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND Sleep loss may induce endothelial dysfunction, a key factor in cardiovascular risk. We examined the endothelial function during one week of sleep restriction and a recovery period (from 3-to-13 days) in healthy subjects, and its link to autonomic, inflammatory and/or endocrine responses. METHODS 12 men were followed at baseline (B1, 8-h sleep), after 2 (SR2) and 6 (SR6) days of SR (4-h sleep: 02:00-06:00) and after 1 (R1) and 12 (R12) recovery nights (8h sleep). At 10:00, we assessed changes in: arm cutaneous vascular conductance (CVC) induced by local application of methacholine (MCh), cathodal current (CIV) and heat (44°C), finger CVC and skin temperature (Tfi) during local cold exposure (5°C, 20-min) and passive recovery (22°C, 20-min). Blood samples were collected at 08:00. RESULTS Compared with baseline (B1), MCh and heat-induced maximal CVC values (CVC peak) were decreased at SR6 and R1. No effect of SR was observed for Tfi and CVC during immersion whereas these values were lower during passive recovery on SR6 and R1. From SR2 to R12, plasma concentrations of insulin, IGF-1 (total and free) and MCP-1 were significantly increased while those of testosterone and prolactin were decreased. Whole-blood blood mRNA concentrations of TNF-α and IL-1β were higher than B1. No changes in noradrenaline concentrations, heart rate and blood pressure were observed. CONCLUSIONS These results demonstrate that SR reduces endothelial-dependent vasodilatation and local tolerance to cold. This endothelial dysfunction is independent of blood pressure and sympathetic activity but associated with inflammatory and metabolic pathway responses (ClinicalTrials-NCT01989741).
Collapse
Affiliation(s)
- Fabien Sauvet
- Institut de recherche biomédicale des armées(IRBA), Brétigny-sur-Orge, France; Université Paris Descartes, Hôtel Dieu, EA7330 VIFASOM (Vigilance, Fatigue, SOmmeil), Paris, France.
| | - Catherine Drogou
- Institut de recherche biomédicale des armées(IRBA), Brétigny-sur-Orge, France; Université Paris Descartes, Hôtel Dieu, EA7330 VIFASOM (Vigilance, Fatigue, SOmmeil), Paris, France
| | - Clément Bougard
- Institut de recherche biomédicale des armées(IRBA), Brétigny-sur-Orge, France; Université Paris Descartes, Hôtel Dieu, EA7330 VIFASOM (Vigilance, Fatigue, SOmmeil), Paris, France
| | - Pierrick J Arnal
- Institut de recherche biomédicale des armées(IRBA), Brétigny-sur-Orge, France; Université Paris Descartes, Hôtel Dieu, EA7330 VIFASOM (Vigilance, Fatigue, SOmmeil), Paris, France; Université de Lyon, Laboratoire de Physiologie de l'Exercice, Saint Etienne, France
| | - Garance Dispersyn
- Institut de recherche biomédicale des armées(IRBA), Brétigny-sur-Orge, France; Université Paris Descartes, Hôtel Dieu, EA7330 VIFASOM (Vigilance, Fatigue, SOmmeil), Paris, France
| | - Cyprien Bourrilhon
- Institut de recherche biomédicale des armées(IRBA), Brétigny-sur-Orge, France
| | - Arnaud Rabat
- Institut de recherche biomédicale des armées(IRBA), Brétigny-sur-Orge, France; Université Paris Descartes, Hôtel Dieu, EA7330 VIFASOM (Vigilance, Fatigue, SOmmeil), Paris, France
| | - Pascal Van Beers
- Institut de recherche biomédicale des armées(IRBA), Brétigny-sur-Orge, France; Université Paris Descartes, Hôtel Dieu, EA7330 VIFASOM (Vigilance, Fatigue, SOmmeil), Paris, France
| | - Danielle Gomez-Merino
- Université Paris Descartes, Hôtel Dieu, EA7330 VIFASOM (Vigilance, Fatigue, SOmmeil), Paris, France
| | - Brice Faraut
- Université Paris Descartes, Hôtel Dieu, EA7330 VIFASOM (Vigilance, Fatigue, SOmmeil), Paris, France; Centre du Sommeil et de la Vigilance, Hôtel Dieu, APHP, Paris, France
| | - Damien Leger
- Université Paris Descartes, Hôtel Dieu, EA7330 VIFASOM (Vigilance, Fatigue, SOmmeil), Paris, France; Centre du Sommeil et de la Vigilance, Hôtel Dieu, APHP, Paris, France
| | - Mounir Chennaoui
- Institut de recherche biomédicale des armées(IRBA), Brétigny-sur-Orge, France; Université Paris Descartes, Hôtel Dieu, EA7330 VIFASOM (Vigilance, Fatigue, SOmmeil), Paris, France
| |
Collapse
|
46
|
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
|
47
|
Specific insulin/IGF1 hybrid receptor activation assay reveals IGF1 as a more potent ligand than insulin. Sci Rep 2015; 5:7911. [PMID: 25604425 PMCID: PMC4300458 DOI: 10.1038/srep07911] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 12/19/2014] [Indexed: 12/15/2022] Open
Abstract
This novel method enables specific measurement of the activation of hybrid receptors formed between the Insulin Receptor (IR) and the Insulin-like Growth Factor 1 Receptor (IGF1R). These hybrid receptors are present in tissues and cell lines expressing both IR and IGF1R. It is therefore challenging to separate the homodimer and hybrid receptor activation properties. This ELISA method enabled fast and quantitative measurements of activated hybrid receptors. The hybrid receptor specificity is obtained from a combination of two specific antibodies for IGF1R and for an IR tyrosine phosphorylation site. The specificity was shown by immunoprecipitations and Western blot analysis. IR exists as two splice variants; consequently, two splice variants of hybrid receptors can be expressed. It is reported here that both splice variants of insulin/IGF1 receptor hybrids are activated by IGF1 with >20-fold higher potency than insulin.
Collapse
|
48
|
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: 23] [Impact Index Per Article: 2.1] [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
|
49
|
Walker AM, Cubbon RM, Kearney MT. Contemporary treatment strategies for Type 2 diabetes-related macrovascular disease. Expert Rev Endocrinol Metab 2014; 9:641-658. [PMID: 30736201 DOI: 10.1586/17446651.2014.941356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Type 2 diabetes mellitus poses a major challenge to healthcare providers in the coming years as its prevalence increases across the globe. The disease doubles the risk of cardiovascular morbidity and mortality, with 70% of sufferers dying from a cardiac cause. Large clinical trials of current glucose-lowering therapies for Type 2 diabetes have shown no benefit in reducing the risk of macrovascular events. Blood pressure control, angiotensin-converting enzyme inhibitor therapy and improvement of dyslipidemia with statins have proven benefit in reducing cardiovascular risk in Type 2 diabetes. A growing understanding of the importance of pathological processes including endothelial dysfunction, abnormal growth factor biology, oxidative stress, dysregulation of adipokines and deficient vascular repair and regeneration in insulin-resistant states promises new treatments to combat the problem.
Collapse
Affiliation(s)
- Andrew Mn Walker
- a Leeds Multidisciplinary Cardiovascular Research Centre, LIGHT laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, UK
| | - Richard M Cubbon
- a Leeds Multidisciplinary Cardiovascular Research Centre, LIGHT laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, UK
| | | |
Collapse
|
50
|
Differential organ phenotypes after postnatal Igf1r gene conditional deletion induced by tamoxifen in UBC-CreERT2; Igf1r fl/fl double transgenic mice. Transgenic Res 2014; 24:279-94. [DOI: 10.1007/s11248-014-9837-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 09/09/2014] [Indexed: 11/25/2022]
|