Skip Navigation
Skip to contents

Diabetes Metab J : Diabetes & Metabolism Journal

Search
OPEN ACCESS

Articles

Page Path
HOME > Diabetes Metab J > Volume 35(1); 2011 > Article
Review
New Perspectives on Diabetic Vascular Complications: The Loss of Endogenous Protective Factors Induced by Hyperglycemia
In-Kyung Jeong1, George L. King2
Diabetes & Metabolism Journal 2011;35(1):8-11.
DOI: https://doi.org/10.4093/dmj.2011.35.1.8
Published online: February 28, 2011
  • 6,242 Views
  • 33 Download
  • 24 Crossref
  • 28 Scopus

1Department of Endocrinology and Metabolism, Kyung Hee University School of Medicine, Seoul, Korea.

2Section on Vascular Cell Biology, Joslin Diabetes Center, Harvard University, Boston, MA, USA.

Corresponding author: George L. King. Section on Vascular Cell Biology, Joslin Diabetes Center One Joslin Place, Boston, MA 02215, USA. George.King@joslin.harvard.edu

Copyright © 2011 Korean Diabetes Association

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

  • Diabetic vascular complications are among the leading causes of morbidity and mortality in diabetic patients. In the past, many studies have focused on the mechanisms of hyperglycemia-induced chronic vascular complications via the formation of toxic metabolites such as oxidative stress, advanced glycosylated end products, persistent activation of protein kinase C, and increased sorbitol concentrations. However, vascular complications result from imbalances caused by increases in systemic toxic metabolites, such as those that occur under conditions of hyperglycemia and dyslipidemia, and by reductions in endogenous protective factors such as insulin, vascular endothelial growth factor, and platelet derived growth factor. This review outlines some of the evidence supporting the importance of enhancing endogenous regenerative factors.
The vascular complications of diabetes mellitus affect many organ systems, including the retina, kidney, nerve, and cardiovascular system. These serious complications are the leading causes of mortality and morbidity in diabetic patients.
Diabetic vascular complications result from imbalances caused by increases in the toxic effects of systemic metabolic abnormalities such as hyperglycemia, dyslipidemia, and hypertension, and reductions in the regenerative effects of endogenous protective factors such as insulin, vascular endothelial growth factor (VEGF), platelet derived growth factor (PDGF), nitric oxide (NO), and antioxidant enzymes (Fig. 1). In the past, many studies on the mechanisms of diabetic complications have focused on the mechanisms by which hyperglycemia might lead to the chronic vascular complications via the formation of toxic metabolites such as oxidants and advanced glycosylated products. These mechanisms include increases in oxidative stress, persistent activation of protein kinase C (PKC) and other signaling pathways, increased sorbitol concentrations through the aldose reductase pathway, the elevated formation of advanced glycosylation end products, and increased flux through the hexosamine pathway [1]. However, few studies have evaluated the importance of endogenous protective factors or the inhibitory effects of hyperglycemia in neutralizing these protective factors during the initiation and progression of diabetic complications. This review outlines some of the evidence supporting the importance of preventing and delaying the progression of diabetic complications. Further, the lack of success in finding effective clinical therapeutics to neutralize the toxic effects of hyperglycemia could be due to the need for enhancing protective factors.
Hyperglycemia-induced cellular apoptosis is a common pathology of many diabetic complications such as for retinal pericytes, renal podocytes, and vascular endothelial cells. In the case of diabetic retinopathy, accelerated pericyte apoptosis is one of the earliest and most specific findings of diabetic complications. Enge et al. [2] reported that PDGF-B or PDGF receptor-β knockout mice exhibited pericyte apoptosis and retinal microvascular abnormalities similar to the early stages of diabetic retinopathy, indicating that PDGF-B is a very important survival factor for retinal pericytes. However, the level of PDGF-B expression was observed to be elevated in diabetic state compared with that in non-diabetic animal [3]. Thus, it was not clear whether pericyte loss results from PDGF-B abundance or PDGF-B resistance. Recently, Geraldes et al. [4] clearly demonstrated that hyperglycemia induced a persistent activation of PKC-δ, which leads to PDGF resistance in the retina. Under normal glucose condition, PDGF-B stimulated DNA synthesis, inhibited cellular apoptosis, and increased p-AKT and p-ERK activation in retinal pericytes. However, PDGF-induced activation of p-AKT and p-ERK signaling was blunted by hyperglycemic levels, which was restored in cells expressing dominant negative PKC-δ or in PKC-δ knockout mice. Geraldes et al. also identified SHP-1, a tyrosine kinase, as a cellular target of PKC-δ and p38α that inhibited the survival action of tyrosine kinase growth factor receptors such as PDGFR-β, by an NF-KB-independent pathway. Therefore, hyperglycemia induces activation of PKC-δ - p38α MAPK - SHP-1, which leads to the inhibition of PDGF-related survival actions that cause pericyte apoptosis in diabetic retinopathy. This study has identified SHP-1 as a new potential therapeutic target, to be inhibited as a treatment for diabetic vascular complications.
Another survival factor is VEGF, which is one of the most important endogenous angiogenic polypeptides that respond to hypoxia under normal physiological conditions. The expression of VEGF is increased in the retina, causing diabetic proliferative retinopathy; however, its abnormally low expression in the myocardium and peripheral lesions causes poor collateral vessel formation in peripheral tissues. During angiogenesis, VEGF interacts with several other angiogenic factors, playing an important role in cell proliferation, differentiation, migration, cell survival, NO production, release of other growth factors, and sympathetic innervation. VEGF may have actions in non-endothelial cells. VEGF is also produced by renal podocytes. Podocytes and fenestrated endothelial cells are two major cells of the glomerular filtration barrier. Mesangial cells are located between the capillary loops and provide structural support. The apoptosis of podocytes is an early event of diabetic nephropathy and a likely cause of proteinuria. Sison et al. [5] reported that VEGF-A released by podocytes was required for glomerular endothelial cell migration, differentiation, and survival through cell-specific gene targeting. The tyrosine kinase receptors for VEGF (VEGFR-1 and VEGFR-2) were expressed by glomerular endothelial cells. Their podocyte-specific, heterozygous, conditional knock out of VEGF-A mice showed a loss of endothelial cell fenestrations, endothelial cell necrosis, loss of podocyte foot process, and a marked loss of mesangial cells. This suggests that the paracrine effects of VEGF are crucial for glomerulus development and maintaining the function of the glomerular filtration barrier.
Microalbuminuria is an early detectable clinical abnormality in diabetic glomerulopathy. Metabolic pathways activated by hyperglycemia, glycated proteins, hemodynamic factors, and oxidative stress are key players in the pathogenesis of diabetic nephropathy. A variety of growth factors and cytokines are then induced through complex signal transduction pathways. Podocyte-derived VEGF is an angiogenic factor whose expression is increased in animal models of diabetic kidney disease [6]. Controversies exist on the effect of the diabetes-induced enhanced expression of VEGF in the renal glomeruli. Some have suggested that VEGFs act in a novel autocrine signaling mode to induce the podocytopathy of diabetes, especially the genesis of albuminuria. Thus, treatment with anti-VEGF antibodies may attenuate glomerular basement membrane thickening and progression of diabetic nephropathy. However, recently Eremina et al. [7] reported proteinuria, hypertension, and renal failure in several patients treated with anti-VEGF agent, bevacizumab. Pathologic findings from kidney biopsies were prominent endothelial swelling, red cell fragments, mesangiolysis, and thrombi in capillary loops which were similar to those in podocyte-specific VEGF-A knockout mice. The study also suggested that anti-VEGF therapy inhibits the VEGF action of podocytes and results in endothelial damage and thrombotic microangiopathy.
An alternative explanation is that the increased VEGF expression in the kidney is a compensatory action to protect the glomeruli from the toxic effects of hyperglycemia. This means that the increase of VEGF is a compensatory response to overcome ischemic or other stressful injury which can induce VEGF resistance in the renal glomeruli by diabetes.
In contrast with diabetic microvascular complications, VEGF expression is reduced in diabetic heart and macrovasculature. Ventricular tissues from muscle insulin receptor knockout mice, which lack insulin receptors in the myocardium, have significant reductions in insulin but not IGF-I signaling, VEGF expression, and vascular density before and after ischemia versus controls [8]. This suggests that impaired insulin signaling has decreased VEGF expression and aggravated ischemic injury. Thus, VEGF is an important angiogenic and survival cytokine in protecting micro- and macro-vascular tissues from conditions such as hyperglycemia.
Insulin is another important survival factor for vascular endothelial cells. Insulin has been shown to have both anti-atherosclerotic and pro-atherosclerotic actions in vascular cells. The anti-atherosclerotic actions of insulin include decreased endothelial cell apoptosis and reactive oxygen species, and increased NO by activation of endothelial NO synthase and heme oxygenase-1. In contrast, insulin has also been reported to have pro-atherosclerotic actions such as increasing endothelin-1 and plasminogen activator inhibitor-1, and enhancing the migration and growth of vascular smooth muscle cells. In diabetes, hyperglycemia can selectively inhibit insulin's anti-atherosclerotic actions, while hyperinsulinemia, as observed in insulin-resistant type 2 diabetes mellitus, can enhance insulin's pro-atherosclerotic effects to accelerate the prevalence of atherosclerotic diseases [9]. Rask-Madsen et al. [10] recently reported that loss of insulin signaling in endothelial cells accelerated atherosclerosis in apolipoprotein E-null mice. Double knockout mice with a deletion of the endothelial insulin receptor and apolipoprotein E gene (EIRAKO) were created using the Cre/loxP system to achieve tissue-specific deletion of these genes. Despite retaining systemic insulin sensitivity, glucose tolerance, plasma lipids, and blood pressure were not different between the EIRAKO and control mice; however, more severe atherosclerotic lesions, impaired vasodilatation, and increased VCAM-1 expression were observed in the EIRAKO mice vs. the apolipoprotein E-null mice. Therefore, improving insulin sensitivity in the endothelium may decrease the risk for atherosclerosis in insulin resistance and diabetes states.
Clinical evidences are also available in support of the importance of endogenous protective factors. Recently, we reported that a significant number of type 1 diabetes mellitus patients with a duration of diabetes of 50 years or longer do not have significant retinopathy and nephropathy (Joslin Medalist Study) [11]. This study demonstrated a lack of association between degree of hyperglycemia and prevalence of microvascular complications, although as a group, glycemic control in the study participants is very good, with a mean HbA1c of 7.3%. These results suggest that unknown endogenous protective factors are greatly reducing microvascular complications in this group of type 1 diabetic subjects of extremely long duration. If the protective factors could be found from the study of these patients, we may be able to prevent or delay microvascular complications in all diabetic patients.
In summary, new therapeutic approaches for diabetic vascular complications need to focus on decreasing the toxic factors of diabetes, but also need to focus on increasing the action of protective factors.
  • 1. Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature 2001;414:813-820. ArticlePubMedPDF
  • 2. Enge M, Bjarnegard M, Gerhardt H, Gustafsson E, Kalen M, Asker N, Hammes HP, Shani M, Fassler R, Betsholtz C. Endothelium-specific platelet-derived growth factor-B ablation mimics diabetic retinopathy. EMBO J 2002;21:4307-4316. ArticlePubMedPMC
  • 3. Yokota T, Ma RC, Park JY, Isshiki K, Sotiropoulos KB, Rauniyar RK, Bornfeldt KE, King GL. Role of protein kinase C on the expression of platelet-derived growth factor and endothelin-1 in the retina of diabetic rats and cultured retinal capillary pericytes. Diabetes 2003;52:838-845. ArticlePubMedPDF
  • 4. Geraldes P, Hiraoka-Yamamoto J, Matsumoto M, Clermont A, Leitges M, Marette A, Aiello LP, Kern TS, King GL. Activation of PKC-delta and SHP-1 by hyperglycemia causes vascular cell apoptosis and diabetic retinopathy. Nat Med 2009;15:1298-1306. ArticlePubMedPMCPDF
  • 5. Sison K, Eremina V, Baelde H, Min W, Hirashima M, Fantus IG, Quaggin SE. Glomerular structure and function require paracrine, not autocrine, VEGF-VEGFR-2 signaling. J Am Soc Nephrol 2010;21:1691-1701. ArticlePubMedPMC
  • 6. Ziyadeh FN. Different roles for TGF-beta and VEGF in the pathogenesis of the cardinal features of diabetic nephropathy. Diabetes Res Clin Pract 2008;82(Suppl 1):S38-S41. PubMed
  • 7. Eremina V, Jefferson JA, Kowalewska J, Hochster H, Haas M, Weisstuch J, Richardson C, Kopp JB, Kabir MG, Backx PH, Gerber HP, Ferrara N, Barisoni L, Alpers CE, Quaggin SE. VEGF inhibition and renal thrombotic microangiopathy. N Engl J Med 2008;358:1129-1136. ArticlePubMedPMC
  • 8. He Z, Opland DM, Way KJ, Ueki K, Bodyak N, Kang PM, Izumo S, Kulkarni RN, Wang B, Liao R, Kahn CR, King GL. Regulation of vascular endothelial growth factor expression and vascularization in the myocardium by insulin receptor and PI3K/Akt pathways in insulin resistance and ischemia. Arterioscler Thromb Vasc Biol 2006;26:787-793. ArticlePubMed
  • 9. Rask-Madsen C, King GL. Mechanisms of disease: endothelial dysfunction in insulin resistance and diabetes. Nat Clin Pract Endocrinol Metab 2007;3:46-56. ArticlePubMedPDF
  • 10. Rask-Madsen C, Li Q, Freund B, Feather D, Abramov R, Wu IH, Chen K, Yamamoto-Hiraoka J, Goldenbogen J, Sotiropoulos KB, Clermont A, Geraldes P, Dall'Osso C, Wagers AJ, Huang PL, Rekhter M, Scalia R, Kahn CR, King GL. Loss of insulin signaling in vascular endothelial cells accelerates atherosclerosis in apolipoprotein E null mice. Cell Metab 2010;11:379-389. ArticlePubMedPMC
  • 11. Keenan HA, Costacou T, Sun JK, Doria A, Cavellerano J, Coney J, Orchard TJ, Aiello LP, King GL. Clinical factors associated with resistance to microvascular complications in diabetic patients of extreme disease duration: the 50-year medalist study. Diabetes Care 2007;30:1995-1997. PubMed
Fig. 1
Diabetes induces an imbalance between toxic and protective factors to cause complications. FFA, free fatty acid; AGE, advanced glycosylated end product; ROS, reactive oxygen species; PKC, protein kinase C; PDGF, platelet-derived growth factor; VEGF, vascular endothelial growth factor; APC, activated protein C.
dmj-35-8-g001.jpg

Figure & Data

References

    Citations

    Citations to this article as recorded by  
    • Role of platelet-derived growth factor c on endothelial dysfunction in cardiovascular diseases
      Adriana Grismaldo, Luis Sobrevia, Ludis Morales
      Biochimica et Biophysica Acta (BBA) - General Subjects.2022; 1866(10): 130188.     CrossRef
    • Ginkgo biloba extracts protect human retinal Müller glial cells from t-BHP induced oxidative damage by activating the AMPK-Nrf2-NQO-1 axis
      Yue Li, Ke Wang, Xue Zhu, Zhengqi Cheng, Ling Zhu, Michael Murray, Fanfan Zhou
      Journal of Pharmacy and Pharmacology.2022;[Epub]     CrossRef
    • Organic Isothiocyanates as Hydrogen Sulfide Donors
      Alma Martelli, Valentina Citi, Lara Testai, Simone Brogi, Vincenzo Calderone
      Antioxidants & Redox Signaling.2020; 32(2): 110.     CrossRef
    • PGC‐1α, a potential therapeutic target against kidney aging
      Gayoung Lee, Md Jamal Uddin, Yoojeong Kim, Minji Ko, Inyoung Yu, Hunjoo Ha
      Aging Cell.2019;[Epub]     CrossRef
    • Glucometabolic characteristics and higher vascular complication risk in Korean patients with type 2 diabetes with non-albumin proteinuria
      Yongin Cho, Yong-ho Lee, Eun Seok Kang, Bong-soo Cha, Byung-wan Lee
      Journal of Diabetes and its Complications.2019; 33(8): 585.     CrossRef
    • Ameliorative effects of taurine against diabetes: a review
      Inam-u-llah, Fengyuan Piao, Rana Muhammad Aadil, Raheel Suleman, Kaixin Li, Mengren Zhang, Pingan Wu, Muhammad Shahbaz, Zulfiqar Ahmed
      Amino Acids.2018; 50(5): 487.     CrossRef
    • Notoginsenoside Fc attenuates high glucose-induced vascular endothelial cell injury via upregulation of PPAR-γ in diabetic Sprague–Dawley rats
      Jingjing Liu, Chunyu Jiang, Xu Ma, Jianbo Wang
      Vascular Pharmacology.2018; 109: 27.     CrossRef
    • Grape seed proanthocyanidin extract protects the retina against early diabetic injury by activating the Nrf2 pathway
      YAN SUN, CAIMEI XIU, WEI LIU, YUAN TAO, JIANRONG WANG, YI QU
      Experimental and Therapeutic Medicine.2016; 11(4): 1253.     CrossRef
    • Hydrogen peroxide prevents vascular calcification induced ROS production by regulating Nrf-2 pathway
      Wensong Zhang, Yi Li, Hanlu Ding, Yaqin Du, Li Wang
      Renal Failure.2016; 38(7): 1099.     CrossRef
    • TNFSF15 Inhibits Blood Retinal Barrier Breakdown Induced by Diabetes
      Feng Jiang, Qingzhong Chen, Liming Huang, Ying Wang, Zhuhong Zhang, Xiangda Meng, Yuanyuan Liu, Chunjie Mao, Fang Zheng, Jingkai Zhang, Hua Yan
      International Journal of Molecular Sciences.2016; 17(5): 615.     CrossRef
    • Effect of Triflusal on Primary Vascular Dysregulation Compared with Aspirin: A Double-Blind, Randomized, Crossover Trial
      Sanghoon Shin, Kwang-Joon Kim, In-Jeong Cho, Geu-Ru Hong, Yangsoo Jang, Namsik Chung, Young Min Rah, Hyuk-Jae Chang
      Yonsei Medical Journal.2015; 56(5): 1227.     CrossRef
    • NRF2 plays a protective role in diabetic retinopathy in mice
      Zhenhua Xu, Yanhong Wei, Junsong Gong, Hongkwan Cho, James K. Park, Ee-Rah Sung, Hu Huang, Lijuan Wu, Charles Eberhart, James T. Handa, Yunpeng Du, Timothy S. Kern, Rajesh Thimmulappa, Alistair J. Barber, Shyam Biswal, Elia J. Duh
      Diabetologia.2014; 57(1): 204.     CrossRef
    • Inhibition of JNK by compound C66 prevents pathological changes of the aorta in STZ‐induced diabetes
      Yucheng Liu, Yonggang Wang, Xiao Miao, Shanshan Zhou, Yi Tan, Guang Liang, Yang Zheng, Quan Liu, Jian Sun, Lu Cai
      Journal of Cellular and Molecular Medicine.2014; 18(6): 1203.     CrossRef
    • Sulforaphane Attenuation of Type 2 Diabetes-Induced Aortic Damage Was Associated with the Upregulation of Nrf2 Expression and Function
      Yonggang Wang, Zhiguo Zhang, Wanqing Sun, Yi Tan, Yucheng Liu, Yang Zheng, Quan Liu, Lu Cai, Jian Sun
      Oxidative Medicine and Cellular Longevity.2014; 2014: 1.     CrossRef
    • Therapeutic Effect of MG132 on the Aortic Oxidative Damage and Inflammatory Response in OVE26 Type 1 Diabetic Mice
      Xiao Miao, Wenpeng Cui, Weixia Sun, Ying Xin, Bo Wang, Yi Tan, Lu Cai, Lining Miao, Yaowen Fu, Guanfang Su, Yuehui Wang
      Oxidative Medicine and Cellular Longevity.2013; 2013: 1.     CrossRef
    • Risk Factors for the Progression of Intima-Media Thickness of Carotid Arteries: A 2-Year Follow-Up Study in Patients with Newly Diagnosed Type 2 Diabetes
      Sang Ouk Chin, Jin Kyung Hwang, Sang Youl Rhee, Suk Chon, You-Cheol Hwang, Seungjoon Oh, Kyu Jeung Ahn, Ho Yeon Chung, Jeong-taek Woo, Sung-Woon Kim, Young Seol Kim, Ja-Heon Kang, In-Kyung Jeong
      Diabetes & Metabolism Journal.2013; 37(5): 365.     CrossRef
    • Downregulation of Nrf2 and HO-1 expression contributes to oxidative stress in type 2 diabetes mellitus: A study in Juana Koslay City, San Luis, Argentina
      Susana Siewert, Irma González, Lucas Santillán, Roberto Lucero, Marta S. Ojeda, María S. Gimenez
      Journal of Diabetes Mellitus.2013; 03(02): 71.     CrossRef
    • The effect of oral prostaglandin analogue on painful diabetic neuropathy: a double‐blind, randomized, controlled trial
      S. Shin, K. J. Kim, H. ‐J. Chang, B. ‐W. Lee, W. ‐I. Yang, B. ‐S. Cha, D. Choi
      Diabetes, Obesity and Metabolism.2013; 15(2): 185.     CrossRef
    • Areca nut procyanidins ameliorate streptozocin-induced hyperglycemia by regulating gluconeogenesis
      Pei-Ling Huang, Chin-Wen Chi, Tsung-Yun Liu
      Food and Chemical Toxicology.2013; 55: 137.     CrossRef
    • Serum glycated albumin predicts the progression of carotid arterial atherosclerosis
      Sun Ok Song, Kwang Joon Kim, Byung-Wan Lee, Eun Seok Kang, Bong Soo Cha, Hyun Chul Lee
      Atherosclerosis.2012; 225(2): 450.     CrossRef
    • Sulforaphane prevention of diabetes-induced aortic damage was associated with the up-regulation of Nrf2 and its down-stream antioxidants
      Xiao Miao, Yang Bai, Weixia Sun, Wenpeng Cui, Ying Xin, Yuehui Wang, Yi Tan, Lining Miao, Yaowen Fu, Guanfang Su, Lu Cai
      Nutrition & Metabolism.2012;[Epub]     CrossRef
    • Association between EPCs count and rate of coronary revascularization in asymptomatic type 2 diabetic patients
      Hyun Min Kim, Kwang Joon Kim, Jae Hoon Moon, Hye-Jeong Lee, Min Kyung Chae, Hyuk-Jae Chang, Eun Seok Kang, Bong Soo Cha, Hyun Chul Lee, Young Jin Kim, Byung-Wan Lee
      Acta Diabetologica.2012; 49(6): 413.     CrossRef
    • Taurine ameliorates hyperglycemia and dyslipidemia by reducing insulin resistance and leptin level in Otsuka Long-Evans Tokushima fatty (OLETF) rats with long-term diabetes
      Kyoung Soo Kim, Da Hee Oh, Jung Yeon Kim, Bong Gn Lee, Jeong Soon You, Kyung Ja Chang, Hyunju Chung, Myung Chul Yoo, Hyung-In Yang, Ja-Heon Kang, Yoo Chul Hwang, Kue Jeong Ahn, Ho-Yeon Chung, In-Kyung Jeong
      Experimental & Molecular Medicine.2012; 44(11): 665.     CrossRef
    • Inhibition of NF-κB prevents high glucose-induced proliferation and plasminogen activator inhibitor-1 expression in vascular smooth muscle cells
      In-Kyung Jeong, Da Hee Oh, Seung-Joon Park, Ja-Heon Kang, Sunshin Kim, Myung-Shik Lee, Myung-Jun Kim, Yoo-Chul Hwang, Kyu Jeong Ahn, Ho-Yeon Chung, Min-Kyung Chae, Hyung-Joon Yoo
      Experimental and Molecular Medicine.2011; 43(12): 684.     CrossRef

    • PubReader PubReader
    • Cite
      CITE
      export Copy
      Close
      Download Citation
      Download a citation file in RIS format that can be imported by all major citation management software, including EndNote, ProCite, RefWorks, and Reference Manager.

      Format:
      • RIS — For EndNote, ProCite, RefWorks, and most other reference management software
      • BibTeX — For JabRef, BibDesk, and other BibTeX-specific software
      Include:
      • Citation for the content below
      New Perspectives on Diabetic Vascular Complications: The Loss of Endogenous Protective Factors Induced by Hyperglycemia
      Diabetes Metab J. 2011;35(1):8-11.   Published online February 28, 2011
      Close
    • XML DownloadXML Download
    Figure

    Diabetes Metab J : Diabetes & Metabolism Journal