Skip Navigation
Skip to contents

Diabetes Metab J : Diabetes & Metabolism Journal

Search
OPEN ACCESS

Search

Page Path
HOME > Search
5 "β-cell"
Filter
Filter
Article category
Keywords
Publication year
Authors
Original Article
Normal Glucose Tolerance with a High 1-Hour Postload Plasma Glucose Level Exhibits Decreased β-Cell Function Similar to Impaired Glucose Tolerance
Tae Jung Oh, Se Hee Min, Chang Ho Ahn, Eun Ky Kim, Soo Heon Kwak, Hye Seung Jung, Kyong Soo Park, Young Min Cho
Diabetes Metab J. 2015;39(2):147-153.   Published online March 9, 2015
DOI: https://doi.org/10.4093/dmj.2015.39.2.147
  • 4,143 View
  • 43 Download
  • 14 Web of Science
  • 13 Crossref
AbstractAbstract PDFPubReader   
Background

Subjects with normal glucose tolerance (NGT) who have a high 1-hour postload plasma glucose level (≥155 mg/dL; NGT 1 hour-high) have been shown to be at higher risk for type 2 diabetes than subjects with NGT 1 hour-low postload plasma glucose level (<155 mg/dL). We compared β-cell function in subjects with NGT 1 hour-high, NGT 1 hour-low, and impaired glucose tolerance (IGT).

Methods

We classified subjects into NGT 1 hour-low (n=149), NGT 1 hour-high (n=43), and IGT (n=52). The β-cell function was assessed based on insulinogenic index (IGI), oral disposition index (DI), and insulin secretion-sensitivity index-2 (ISSI-2).

Results

Insulin sensitivity was comparable between the subjects with NGT 1 hour-high and NGT 1 hour-low. The β-cell function with/without adjusting insulin sensitivity was significantly different among the three groups. The IGI (pmol/mmol) was 116.8±107.3 vs. 64.8±47.8 vs. 65.8±80.6 (P=0.141), oral DI was 3.5±4.2 vs. 1.8±1.4 vs. 1.8±3.1 (P<0.001), and ISSI-2 was 301.2±113.7 vs. 213.2±67.3 vs. 172.5±87.5 (P<0.001) in NGT 1 hour-low, NGT 1 hour-high, and IGT, respectively. Post hoc analyses revealed that oral DI and ISSI-2 were significantly different between NGT 1 hour-low and NGT 1 hour-high but comparable between NGT 1 hour-high and IGT.

Conclusion

Among Korean subjects with NGT, those who have a higher 1-hour postload glucose level have a compromised insulin-sensitivity adjusted β-cell function to a similar degree as IGT subjects.

Citations

Citations to this article as recorded by  
  • Triglyceride-glucose index predicts type 2 diabetes mellitus more effectively than oral glucose tolerance test-derived insulin sensitivity and secretion markers
    Min Jin Lee, Ji Hyun Bae, Ah Reum Khang, Dongwon Yi, Mi Sook Yun, Yang Ho Kang
    Diabetes Research and Clinical Practice.2024; 210: 111640.     CrossRef
  • Pancreatic fat accumulation is associated with decreased β‐cell function and deterioration in glucose tolerance in Korean adults
    Sang Ouk Chin, You‐Cheol Hwang, In‐Jin Cho, In‐Kyung Jeong, Kyu Jeung Ahn, Ho Yeon Chung
    Diabetes/Metabolism Research and Reviews.2021;[Epub]     CrossRef
  • Indirect insulin resistance detection: Current clinical trends and laboratory limitations
    Sylwia Placzkowska, Lilla Pawlik-Sobecka, Izabela Kokot, Agnieszka Piwowar
    Biomedical Papers.2019; 163(3): 187.     CrossRef
  • Clinical Implications of Using Post-Challenge Plasma Glucose Levels for Early Diagnosis of Type 2 Diabetes Mellitus in Older Individuals
    Kyong Hye Joung, Sang Hyun Ju, Ji Min Kim, Sorim Choung, Jae Min Lee, Kang Seo Park, Hyun Jin Kim, Bon Jeong Ku
    Diabetes & Metabolism Journal.2018; 42(2): 147.     CrossRef
  • The 1-h post-load plasma glucose as a novel biomarker for diagnosing dysglycemia
    Ram Jagannathan, Martin Buysschaert, José Luis Medina, Karin Katz, Sarah Musleh, Brenda Dorcely, Michael Bergman
    Acta Diabetologica.2018; 55(6): 519.     CrossRef
  • Elevated 1‐hour post‐load plasma glucose identifies obese youth with abnormal glucose metabolism and an unfavourable inflammatory profile
    Anastasios Serbis, Vasileios Giapros, Anna Challa, Nikolaos Chaliasos, Ekaterini Siomou
    Clinical Endocrinology.2018; 89(6): 757.     CrossRef
  • One‐hour postload plasma glucose concentration in people with normal glucose homeostasis predicts future diabetes mellitus: a 12‐year community‐based cohort study
    Tae Jung Oh, Soo Lim, Kyoung Min Kim, Jae Hoon Moon, Sung Hee Choi, Young Min Cho, Kyong Soo Park, HakChul Jang, Nam H. Cho
    Clinical Endocrinology.2017; 86(4): 513.     CrossRef
  • An elevated 1-h post- load glucose level during the oral glucose tolerance test detects prediabetes
    Martin Buysschaert, Michael Bergman, Donald Yanogo, Ram Jagannathan, Benoit Buysschaert, Vanessa Preumont
    Diabetes & Metabolic Syndrome: Clinical Research & Reviews.2017; 11(2): 137.     CrossRef
  • Delayed insulin secretion response during an OGTT is associated with an increased risk for incidence of diabetes in NGT subjects
    Yun Sun, Junfeng Han, Ziwei Lin, Lige Song, Chen Wang, Weiping Jia
    Journal of Diabetes and its Complications.2016; 30(8): 1537.     CrossRef
  • Postprandial Hyperglycemia
    Tae Jung Oh
    The Journal of Korean Diabetes.2016; 17(4): 233.     CrossRef
  • β-Cell Function and Insulin Sensitivity in Normal Glucose-Tolerant Subjects Stratified by 1-Hour Plasma Glucose Values
    Miranda M. Priya, Anandakumar Amutha, T.A. Pramodkumar, Harish Ranjani, Saravanan Jebarani, Kuppan Gokulakrishnan, Rajendra Pradeepa, Ranjit Unnikrishnan, Ranjit Mohan Anjana, Viswanathan Mohan
    Diabetes Technology & Therapeutics.2016; 18(1): 29.     CrossRef
  • Response: Normal Glucose Tolerance with a High 1-Hour Postload Plasma Glucose Level Exhibits Decreased β-Cell Function Similar to Impaired Glucose Tolerance (Diabetes Metab J2015;39:147-53)
    Tae Jung Oh, Se Hee Min, Chang Ho Ahn, Eun Ky Kim, Soo Heon Kwak, Hye Seung Jung, Kyong Soo Park, Young Min Cho
    Diabetes & Metabolism Journal.2015; 39(3): 270.     CrossRef
  • Letter: Normal Glucose Tolerance with a High 1-Hour Postload Plasma Glucose Level Exhibits Decreased β-Cell Function Similar to Impaired Glucose Tolerance (Diabetes Metab J2015;39:147-53)
    Hee Kyung Kim
    Diabetes & Metabolism Journal.2015; 39(3): 268.     CrossRef
Reviews
Therapeutic Approaches for Preserving or Restoring Pancreatic β-Cell Function and Mass
Kyong Yeun Jung, Kyoung Min Kim, Soo Lim
Diabetes Metab J. 2014;38(6):426-436.   Published online December 15, 2014
DOI: https://doi.org/10.4093/dmj.2014.38.6.426
  • 6,144 View
  • 115 Download
  • 20 Web of Science
  • 19 Crossref
AbstractAbstract PDFPubReader   

The goal for the treatment of patients with diabetes has today shifted from merely reducing glucose concentrations to preventing the natural decline in β-cell function and delay the progression of disease. Pancreatic β-cell dysfunction and decreased β-cell mass are crucial in the development of diabetes. The β-cell defects are the main pathogenesis in patients with type 1 diabetes and are associated with type 2 diabetes as the disease progresses. Recent studies suggest that human pancreatic β-cells have a capacity for increased proliferation according to increased demands for insulin. In humans, β-cell mass has been shown to increase in patients showing insulin-resistance states such as obesity or in pregnancy. This capacity might be useful for identifying new therapeutic strategies to reestablish a functional β-cell mass. In this context, therapeutic approaches designed to increase β-cell mass might prove a significant way to manage diabetes and prevent its progression. This review describes the various β-cell defects that appear in patients with diabetes and outline the mechanisms of β-cell failure. We also review common methods for assessing β-cell function and mass and methodological limitations in vivo. Finally, we discuss the current therapeutic approaches to improve β-cell function and increase β-cell mass.

Citations

Citations to this article as recorded by  
  • Polychlorinated biphenyls exposure and type 2 diabetes: Molecular mechanism that causes insulin resistance and islet damage
    Qiuli Shan, Jingyu Liu, Fan Qu, Anhui Chen, Wenxing He
    Environmental Toxicology.2024; 39(4): 2466.     CrossRef
  • Beta‐cell function in type 2 diabetes (T2DM): Can it be preserved or enhanced?
    Laure Sayyed Kassem, Aman Rajpal, Margarita Victoria Barreiro, Faramarz Ismail‐Beigi
    Journal of Diabetes.2023; 15(10): 817.     CrossRef
  • Preliminary Evaluation of Potential Properties of Three Probiotics and Their Combination with Prebiotics on GLP-1 Secretion and Type 2 Diabetes Alleviation
    Ran Xiao, Ran Wang, Shusen Li, Xiaohong Kang, Yimei Ren, Erna Sun, Chenyuan Wang, Jingjing He, Jing Zhan, Wen Yi Kang
    Journal of Food Quality.2022; 2022: 1.     CrossRef
  • Preservation effect of imeglimin on pancreatic β-cell mass: Noninvasive evaluation using 111In-exendin-4 SPECT/CT imaging and the perspective of mitochondrial involvements
    Muhammad Fauzi, Takaaki Murakami, Hiroyuki Fujimoto, Ainur Botagarova, Kentaro Sakaki, Sakura Kiyobayashi, Masahito Ogura, Nobuya Inagaki
    Frontiers in Endocrinology.2022;[Epub]     CrossRef
  • Effects of boschnaloside from Boschniakia rossica on dysglycemia and islet dysfunction in severely diabetic mice through modulating the action of glucagon-like peptide-1
    Lie-Chwen Lin, Lin-Chien Lee, Cheng Huang, Chiung-Tong Chen, Jen-Shin Song, Young-Ji Shiao, Hui-Kang Liu
    Phytomedicine.2019; 62: 152946.     CrossRef
  • The Potential of Hibiscus sabdariffa Linn in Inducing Glucagon-Like Peptide-1 via SGLT-1 and GLPR in DM Rats
    Neng Tine Kartinah, Fadilah Fadilah, Ermita Ilyas Ibrahim, Yuliana Suryati
    BioMed Research International.2019; 2019: 1.     CrossRef
  • Efficacy of short‑term intensive treatment with insulin pump to improve islet β‑cell function in newly diagnosed type 2 diabetes via inhibition of oxidative stress
    Hai‑Tong Liu, Yan Gao
    Experimental and Therapeutic Medicine.2019;[Epub]     CrossRef
  • When metformin is not enough: Pros and cons of SGLT2 and DPP‐4 inhibitors as a second line therapy
    Angelo Avogaro, Elías Delgado, Ildiko Lingvay
    Diabetes/Metabolism Research and Reviews.2018;[Epub]     CrossRef
  • Metformin prevents glucotoxicity by alleviating oxidative and ER stress–induced CD36 expression in pancreatic beta cells
    Jun Sung Moon, Udayakumar Karunakaran, Suma Elumalai, In-Kyu Lee, Hyoung Woo Lee, Yong-Woon Kim, Kyu Chang Won
    Journal of Diabetes and its Complications.2017; 31(1): 21.     CrossRef
  • The variability in beta‐cell function in placebo‐treated subjects with type 2 diabetes: application of the weight‐HbA1c‐insulin‐glucose (WHIG) model
    Janna K. Duong, Willem de Winter, Steve Choy, Nele Plock, Himanshu Naik, Walter Krauwinkel, Sandra A.G. Visser, Katia M. Verhamme, Miriam C. Sturkenboom, B.H. Stricker, Meindert Danhof
    British Journal of Clinical Pharmacology.2017; 83(3): 487.     CrossRef
  • Regulation of pancreatic β-cell function and mass dynamics by prostaglandin signaling
    Bethany A. Carboneau, Richard M. Breyer, Maureen Gannon
    Journal of Cell Communication and Signaling.2017; 11(2): 105.     CrossRef
  • Effects of low intensity laser acupoint irradiation on inhibiting islet beta-cell apoptosis in rats with type 2 diabetes
    Guoxin Xiong, Leilei Xiong, Xinzhong Li
    Laser Physics.2016; 26(9): 095605.     CrossRef
  • Effects of continuous positive airway pressure treatment on glucose metabolism in patients with obstructive sleep apnea
    Elisabet Martínez-Ceron, Isabel Fernández-Navarro, Francisco Garcia-Rio
    Sleep Medicine Reviews.2016; 25: 121.     CrossRef
  • Efficacy and safety of teneligliptin, a novel dipeptidyl peptidase‐4 inhibitor, in Korean patients with type 2 diabetes mellitus: a 24‐week multicentre, randomized, double‐blind, placebo‐controlled phase III trial
    S. Hong, C.‐Y. Park, K. A. Han, C. H. Chung, B. J. Ku, H. C. Jang, C. W. Ahn, M.‐K. Lee, M. K. Moon, H. S. Son, C. B. Lee, Y.‐W. Cho, S.‐W. Park
    Diabetes, Obesity and Metabolism.2016; 18(5): 528.     CrossRef
  • Senp2 expression was induced by chronic glucose stimulation in INS1 cells, and it was required for the associated induction of Ccnd1 and Mafa
    Hye Seung Jung, Yu Mi Kang, Ho Seon Park, Byung Yong Ahn, Hakmo Lee, Min Joo Kim, Jin Young Jang, Sun-Whe Kim
    Islets.2016; 8(6): 207.     CrossRef
  • ANTIDIABETIC EVALUATION OF CHONEMORPHA MACROPHYLLA ROOTS BY IN VIVO METHODS USING MALE WISTAR ALBINO RATS AND ITS MECHANISM OF ACTION
    K. R. Krishna Kumar, K. K. Srinivasan
    INDIAN DRUGS.2016; 53(12): 42.     CrossRef
  • Comparative gene expression profiles in pancreatic islets associated with agouti yellow mutation and PACAP overexpression in mice
    Kazuya Ikeda, Shuhei Tomimoto, Soken Tsuchiya, Ken-ichi Hamagami, Norihito Shintani, Yukihiko Sugimoto, Atsushi Ichikawa, Atsushi Kasai, Takanobu Nakazawa, Kazuki Nagayasu, Atsuko Hayata-Takano, Akemichi Baba, Hitoshi Hashimoto
    Biochemistry and Biophysics Reports.2015; 2: 179.     CrossRef
  • Therapeutic efficacy of umbilical cord-derived mesenchymal stem cells in patients with type 2 diabetes
    LI-XUE GUAN, HUI GUAN, HAI-BO LI, CUI-AI REN, LIN LIU, JIN-JIN CHU, LONG-JUN DAI
    Experimental and Therapeutic Medicine.2015; 9(5): 1623.     CrossRef
  • p13 overexpression in pancreatic β-cells ameliorates type 2 diabetes in high-fat-fed mice
    Shintaro Higashi, Kazuhiko Katagi, Norihito Shintani, Kazuya Ikeda, Yukihiko Sugimoto, Soken Tsuchiya, Naoki Inoue, Shota Tanaka, Mai Koumoto, Atsushi Kasai, Takanobu Nakazawa, Atsuko Hayata-Takano, Ken-Ichi Hamagami, Shuhei Tomimoto, Takuya Yoshida, Tada
    Biochemical and Biophysical Research Communications.2015; 461(4): 612.     CrossRef
Neonatal Diabetes Caused by Activating Mutations in the Sulphonylurea Receptor
Peter Proks
Diabetes Metab J. 2013;37(3):157-164.   Published online June 14, 2013
DOI: https://doi.org/10.4093/dmj.2013.37.3.157
  • 3,511 View
  • 51 Download
  • 10 Crossref
AbstractAbstract PDFPubReader   

Adenosine triphosphate (ATP)-sensitive potassium (KATP) channels in pancreatic β-cells play a crucial role in insulin secretion and glucose homeostasis. These channels are composed of two subunits: a pore-forming subunit (Kir6.2) and a regulatory subunit (sulphonylurea receptor-1). Recent studies identified large number of gain of function mutations in the regulatory subunit of the channel which cause neonatal diabetes. Majority of mutations cause neonatal diabetes alone, however some lead to a severe form of neonatal diabetes with associated neurological complications. This review focuses on the functional effects of these mutations as well as the implications for treatment.

Citations

Citations to this article as recorded by  
  • Long-term Follow-up of Glycemic and Neurological Outcomes in an International Series of Patients With Sulfonylurea-Treated ABCC8 Permanent Neonatal Diabetes
    Pamela Bowman, Frances Mathews, Fabrizio Barbetti, Maggie H. Shepherd, Janine Sanchez, Barbara Piccini, Jacques Beltrand, Lisa R. Letourneau-Freiberg, Michel Polak, Siri Atma W. Greeley, Eamon Rawlins, Tarig Babiker, Nicholas J. Thomas, Elisa De Franco, S
    Diabetes Care.2021; 44(1): 35.     CrossRef
  • Structure based analysis of KATP channel with a DEND syndrome mutation in murine skeletal muscle
    Shoichiro Horita, Tomoyuki Ono, Saul Gonzalez-Resines, Yuko Ono, Megumi Yamachi, Songji Zhao, Carmen Domene, Yuko Maejima, Kenju Shimomura
    Scientific Reports.2021;[Epub]     CrossRef
  • Clinical and Genetic Characteristics of ABCC8 Nonneonatal Diabetes Mellitus: A Systematic Review
    Meng Li, Xueyao Han, Linong Ji, Karim Gariani
    Journal of Diabetes Research.2021; 2021: 1.     CrossRef
  • Spacial models of malfunctioned protein complexes help to elucidate signal transduction critical for insulin release
    Katarzyna Walczewska-Szewc, Wieslaw Nowak
    Biosystems.2019; 177: 48.     CrossRef
  • Cantu syndrome–associated SUR2 (ABCC9) mutations in distinct structural domains result in KATP channel gain-of-function by differential mechanisms
    Conor McClenaghan, Alex Hanson, Monica Sala-Rabanal, Helen I. Roessler, Dragana Josifova, Dorothy K. Grange, Gijs van Haaften, Colin G. Nichols
    Journal of Biological Chemistry.2018; 293(6): 2041.     CrossRef
  • Hyperinsulinism-Causing Mutations Cause Multiple Molecular Defects in SUR1 NBD1
    Claudia P. Alvarez, Marijana Stagljar, D. Ranjith Muhandiram, Voula Kanelis
    Biochemistry.2017; 56(18): 2400.     CrossRef
  • KATP Channel Mutations and Neonatal Diabetes
    Kenju Shimomura, Yuko Maejima
    Internal Medicine.2017; 56(18): 2387.     CrossRef
  • Molecular action of sulphonylureas on KATP channels: a real partnership between drugs and nucleotides
    Heidi de Wet, Peter Proks
    Biochemical Society Transactions.2015; 43(5): 901.     CrossRef
  • Successful development and use of a thermodynamic stability screen for optimizing the yield of nucleotide binding domains
    Elvin D. de Araujo, Voula Kanelis
    Protein Expression and Purification.2014; 103: 38.     CrossRef
  • Reclasificación y transferencia de insulina a sulfonilureas en un paciente con mutación en KCNJ11 tras 15 años de tratamiento con insulina
    Magdalena Capponi, Carmen Quirós, Ignacio Conget, Enric Esmatjes, Marga Giménez
    Avances en Diabetología.2014; 30(4): 115.     CrossRef
The Role of the Sweet Taste Receptor in Enteroendocrine Cells and Pancreatic β-Cells
Itaru Kojima, Yuko Nakagawa
Diabetes Metab J. 2011;35(5):451-457.   Published online October 31, 2011
DOI: https://doi.org/10.4093/dmj.2011.35.5.451
  • 4,359 View
  • 86 Download
  • 45 Crossref
AbstractAbstract PDFPubReader   

The sweet taste receptor is expressed in taste cells located in taste buds of the tongue. This receptor senses sweet substances in the oral cavity, activates taste cells, and transmits the taste signals to adjacent neurons. The sweet taste receptor is a heterodimer of two G protein-coupled receptors, T1R2 and T1R3. Recent studies have shown that this receptor is also expressed in the extragustatory system, including the gastrointestinal tract, pancreatic β-cells, and glucose-responsive neurons in the brain. In the intestine, the sweet taste receptor regulates secretion of incretin hormones and glucose uptake from the lumen. In β-cells, activation of the sweet taste receptor leads to stimulation of insulin secretion. Collectively, the sweet taste receptor plays an important role in recognition and metabolism of energy sources in the body.

Citations

Citations to this article as recorded by  
  • Targeting T2Rs, a feasible approach for natural bitter agents from traditional Chinese medicine modulate ABC transporters to treat respiratory diseases
    Qi Liang, Ruo-Lan Li, Dan-Dan Tang, Ting Zhang, Lian Zhong, Chun-Jie Wu, Wei Peng
    Arabian Journal of Chemistry.2024; 17(1): 105377.     CrossRef
  • Impact of dietary sucralose and sucrose-sweetened water intake on lipid and glucose metabolism in male mice
    Xinyi Wu, Le Cui, Haoquan Wang, Jinhong Xu, Zhaozhao Zhong, Xibei Jia, Jiaqi Wang, Huahua Zhang, Yanteng Shi, Yuhang Tang, Qianhui Yang, Qiongdan Liang, Yujing Zhang, Jing Li, Xiaohong Jiang
    European Journal of Nutrition.2023; 62(1): 199.     CrossRef
  • How dietary amino acids and high protein diets influence insulin secretion
    Yuuki Yanagisawa
    Physiological Reports.2023;[Epub]     CrossRef
  • The elusive cephalic phase insulin response: triggers, mechanisms, and functions
    Wolfgang Langhans, Alan G. Watts, Alan C. Spector
    Physiological Reviews.2023; 103(2): 1423.     CrossRef
  • Trace Amine-Associated Receptors and Monoamine-Mediated Regulation of Insulin Secretion in Pancreatic Islets
    Anastasia N. Vaganova, Taisiia S. Shemyakova, Karina V. Lenskaia, Roman N. Rodionov, Charlotte Steenblock, Raul R. Gainetdinov
    Biomolecules.2023; 13(11): 1618.     CrossRef
  • Gene expression analyses of TAS1R taste receptors relevant to the treatment of cardiometabolic disease
    Mariah R Stavrou, Sean Souchiart So, Angela M Finch, Sara Ballouz, Nicola J Smith
    Chemical Senses.2023;[Epub]     CrossRef
  • Dysgeusia
    Davis C. Thomas, Deepti Chablani, Srishti Parekh, Reshmy Chellam Pichammal, Karpagavalli Shanmugasundaram, Priyanka Kodaganallur Pitchumani
    The Journal of the American Dental Association.2022; 153(3): 251.     CrossRef
  • Oral Microbiota-Host Interaction Mediated by Taste Receptors
    Hao Dong, Jiaxin Liu, Jianhui Zhu, Zhiyan Zhou, Marco Tizzano, Xian Peng, Xuedong Zhou, Xin Xu, Xin Zheng
    Frontiers in Cellular and Infection Microbiology.2022;[Epub]     CrossRef
  • New frontiers in the hunger management involving GLP‐1, taste and oestrogen
    Maja Baretić
    Diabetic Medicine.2022;[Epub]     CrossRef
  • The Impact of Artificial Sweeteners on Body Weight Control and Glucose Homeostasis
    Michelle D. Pang, Gijs H. Goossens, Ellen E. Blaak
    Frontiers in Nutrition.2021;[Epub]     CrossRef
  • TAS1R2 sweet taste receptor genetic variation and dietary intake in Korean females
    Jeong-Hwa Choi
    Appetite.2021; 164: 105281.     CrossRef
  • Sweet Taste Is Complex: Signaling Cascades and Circuits Involved in Sweet Sensation
    Elena von Molitor, Katja Riedel, Michael Krohn, Mathias Hafner, Rüdiger Rudolf, Tiziana Cesetti
    Frontiers in Human Neuroscience.2021;[Epub]     CrossRef
  • Hazardous Hyperglisemic Effect of Facial Ischemia Following Subarachnoid Hemorrhage: An Experimental Study
    Mehmet Dumlu Aydin, Ozgur Caglar, Mehmet Nuri Kocak, Erdem Karadeniz, Nazan Aydin, Irem Ates, Sevilay Ozmen
    Archives of Neuroscience.2020;[Epub]     CrossRef
  • Determinants of Sweetness Preference: A Scoping Review of Human Studies
    Carolina Venditti, Kathy Musa-Veloso, Han Youl Lee, Theresa Poon, Alastair Mak, Maryse Darch, Justine Juana, Dylan Fronda, Daniel Noori, Erika Pateman, Maia Jack
    Nutrients.2020; 12(3): 718.     CrossRef
  • Allelic variation of the Tas1r3 taste receptor gene affects sweet taste responsiveness and metabolism of glucose in F1 mouse hybrids
    Vladimir O. Murovets, Ekaterina A. Lukina, Egor A. Sozontov, Julia V. Andreeva, Raisa P. Khropycheva, Vasiliy A. Zolotarev, Keiko Abe
    PLOS ONE.2020; 15(7): e0235913.     CrossRef
  • Effect of sucralose and aspartame on glucose metabolism and gut hormones
    Samar Y Ahmad, James K Friel, Dylan S Mackay
    Nutrition Reviews.2020; 78(9): 725.     CrossRef
  • Maternal low protein exposure alters glucose tolerance and intestinal nutrient-responsive receptors and transporters expression of rat offspring
    Nan Wang, Bo Lv, Limin Guan, Hu Qiao, Bo Sun, Xiao Luo, Ru Jia, Ke Chen, Jianqun Yan
    Life Sciences.2020; 243: 117216.     CrossRef
  • Sugar Reduction in Dairy Food: An Overview with Flavoured Milk as an Example
    Dipendra Kumar Mahato, Russell Keast, Djin Gie Liem, Catherine Georgina Russell, Sara Cicerale, Shirani Gamlath
    Foods.2020; 9(10): 1400.     CrossRef
  • Electrophysiology of the pancreatic islet β-cell sweet taste receptor TIR3
    Juan V. Sanchez-Andres, Willy J. Malaisse, Itaru Kojima
    Pflügers Archiv - European Journal of Physiology.2019; 471(4): 647.     CrossRef
  • A Single 48 mg Sucralose Sip Unbalances Monocyte Subpopulations and Stimulates Insulin Secretion in Healthy Young Adults
    Angélica Y. Gómez-Arauz, Nallely Bueno-Hernández, Leon F. Palomera, Raúl Alcántara-Suárez, Karen L. De León, Lucía A. Méndez-García, Miguel Carrero-Aguirre, Aaron N. Manjarrez-Reyna, Camilo P. Martínez-Reyes, Marcela Esquivel-Velázquez, Alejandra Ruiz-Bar
    Journal of Immunology Research.2019; 2019: 1.     CrossRef
  • Sugar reduction without compromising sensory perception. An impossible dream?
    Scott C. Hutchings, Julia Y. Q. Low, Russell S. J. Keast
    Critical Reviews in Food Science and Nutrition.2019; 59(14): 2287.     CrossRef
  • Oral and Post‐Oral Actions of Low‐Calorie Sweeteners: A Tale of Contradictions and Controversies
    John I. Glendinning
    Obesity.2018;[Epub]     CrossRef
  • Olfactory, Taste, and Photo Sensory Receptors in Non-sensory Organs: It Just Makes Sense
    Nicholas M. Dalesio, Sebastian F. Barreto Ortiz, Jennifer L. Pluznick, Dan E. Berkowitz
    Frontiers in Physiology.2018;[Epub]     CrossRef
  • Emerging Concepts in Brain Glucose Metabolic Functions: From Glucose Sensing to How the Sweet Taste of Glucose Regulates Its Own Metabolism in Astrocytes and Neurons
    Menizibeya O. Welcome, Nikos E. Mastorakis
    NeuroMolecular Medicine.2018; 20(3): 281.     CrossRef
  • Anti-diabetic effects of natural products an overview of therapeutic strategies
    Jiyoung Park, Hyeung-Jin Jang
    Molecular & Cellular Toxicology.2017; 13(1): 1.     CrossRef
  • Natural sweetener agave inhibits gastric emptying in rats by a cholecystokinin-2- and glucagon like peptide-1 receptor-dependent mechanism
    E. Bihter Gürler, Dilek Özbeyli, Hülya Buzcu, Sezin Bayraktar, İrem Carus, Beyza Dağ, Yasemin Geriş, Seda Jeral, Berrak Ç. Yeğen
    Food & Function.2017; 8(2): 741.     CrossRef
  • T1R3 homomeric sweet taste receptor regulates adipogenesis through Gαs-mediated microtubules disassembly and Rho activation in 3T3-L1 cells
    Yosuke Masubuchi, Yuko Nakagawa, Johan Medina, Masahiro Nagasawa, Itaru Kojima, Mark M. Rasenick, Takeshi Inagaki, Hiroshi Shibata, Hiroaki Matsunami
    PLOS ONE.2017; 12(5): e0176841.     CrossRef
  • Hormonal responses to non-nutritive sweeteners in water and diet soda
    Allison C. Sylvetsky, Rebecca J. Brown, Jenny E. Blau, Mary Walter, Kristina I. Rother
    Nutrition & Metabolism.2016;[Epub]     CrossRef
  • Gastrointestinal defense mechanisms
    Hyder Said, Jonathan D. Kaunitz
    Current Opinion in Gastroenterology.2016; 32(6): 461.     CrossRef
  • Glucose-Sensing Receptor T1R3: A New Signaling Receptor Activated by Glucose in Pancreatic β-Cells
    Itaru Kojima, Yuko Nakagawa, Kunihisa Hamano, Johan Medina, Longfei Li, Masahiro Nagasawa
    Biological & Pharmaceutical Bulletin.2015; 38(5): 674.     CrossRef
  • Impaired Glucose Metabolism in Mice Lacking the Tas1r3 Taste Receptor Gene
    Vladimir O. Murovets, Alexander A. Bachmanov, Vasiliy A. Zolotarev, Hiroaki Matsunami
    PLOS ONE.2015; 10(6): e0130997.     CrossRef
  • Sweet Taste Receptor Signaling Network: Possible Implication for Cognitive Functioning
    Menizibeya O. Welcome, Nikos E. Mastorakis, Vladimir A. Pereverzev
    Neurology Research International.2015; 2015: 1.     CrossRef
  • Expression of the glucose-sensing receptor T1R3 in pancreatic islet: changes in the expression levels in various nutritional and metabolic states
    Anya Medina, Yuko Nakagawa, Jinhui Ma, Longfei Li, Kunihisa Hamano, Toshio Akimoto, Yuzo Ninomiya, Itaru Kojima
    Endocrine Journal.2014; 61(8): 797.     CrossRef
  • Normal Roles for Dietary Fructose in Carbohydrate Metabolism
    Maren Laughlin
    Nutrients.2014; 6(8): 3117.     CrossRef
  • Glucose promotes its own metabolism by acting on the cell-surface glucose-sensing receptor T1R3
    Yuko Nakagawa, Yoshiaki Ohtsu, Masahiro Nagasawa, Hiroshi Shibata, Itaru Kojima
    Endocrine Journal.2014; 61(2): 119.     CrossRef
  • Diverse signaling systems activated by the sweet taste receptor in human GLP-1-secreting cells
    Yoshiaki Ohtsu, Yuko Nakagawa, Masahiro Nagasawa, Shigeki Takeda, Hirokazu Arakawa, Itaru Kojima
    Molecular and Cellular Endocrinology.2014; 394(1-2): 70.     CrossRef
  • Sweet Taste-Sensing Receptors Expressed in Pancreatic β-Cells: Sweet Molecules Act as Biased Agonists
    Itaru Kojima, Yuko Nakagawa, Yoshiaki Ohtsu, Anya Medina, Masahiro Nagasawa
    Endocrinology and Metabolism.2014; 29(1): 12.     CrossRef
  • The Role of Sweet Taste in Satiation and Satiety
    Yu Low, Kathleen Lacy, Russell Keast
    Nutrients.2014; 6(9): 3431.     CrossRef
  • Insulin release: the receptor hypothesis
    Willy J. Malaisse
    Diabetologia.2014; 57(7): 1287.     CrossRef
  • The involvement of the T1R3 receptor protein in the control of glucose metabolism in mice at different levels of glycemia
    V. O. Murovets, A. A. Bachmanov, S. V. Travnikov, A. A. Churikova, V. A. Zolotarev
    Journal of Evolutionary Biochemistry and Physiology.2014; 50(4): 334.     CrossRef
  • GKAs for diabetes therapy: why no clinically useful drug after two decades of trying?
    Franz M. Matschinsky
    Trends in Pharmacological Sciences.2013; 34(2): 90.     CrossRef
  • A Novel Regulatory Function of Sweet Taste-Sensing Receptor in Adipogenic Differentiation of 3T3-L1 Cells
    Yosuke Masubuchi, Yuko Nakagawa, Jinhui Ma, Tsutomu Sasaki, Tadahiro Kitamura, Yoritsuna Yamamoto, Hitoshi Kurose, Itaru Kojima, Hiroshi Shibata, Xing-Ming Shi
    PLoS ONE.2013; 8(1): e54500.     CrossRef
  • Goût des aliments et comportement alimentaire
    M. Fantino
    Médecine des Maladies Métaboliques.2012; 6(5): 409.     CrossRef
  • The role of T1r3 and Trpm5 in carbohydrate-induced obesity in mice
    John I. Glendinning, Jennifer Gillman, Haley Zamer, Robert F. Margolskee, Anthony Sclafani
    Physiology & Behavior.2012; 107(1): 50.     CrossRef
  • Repair of diverse diabetic defects of β‐cells in man and mouse by pharmacological glucokinase activation
    Nicolai M. Doliba, Deborah Fenner, Bogumil Zelent, Joseph Bass, Ramakanth Sarabu, Franz M. Matschinsky
    Diabetes, Obesity and Metabolism.2012; 14(s3): 109.     CrossRef
Glucolipotoxicity in Pancreatic β-Cells
Ji-Won Kim, Kun-Ho Yoon
Diabetes Metab J. 2011;35(5):444-450.   Published online October 31, 2011
DOI: https://doi.org/10.4093/dmj.2011.35.5.444
  • 54,475 View
  • 90 Download
  • 58 Crossref
AbstractAbstract PDFPubReader   

The recent epidemic of type 2 diabetes in Asia differs from that reported in other regions of the world in several key areas: it has evolved over a much shorter time, in an earlier stage of life, and in people with lower body mass indices. These phenotypic characteristics of patients strongly suggest that insulin secretory defects may perform a more important function in the development and progression of diabetes. A genetic element clearly underlies β-cell dysfunction and insufficient β-cell mass; however, a number of modifiable factors are also linked to β-cell deterioration, most notably chronic hyperglycemia and elevated free fatty acid (FFA) levels. Neither glucose nor FFAs alone cause clinically meaningful β-cell toxicity, especially in patients with normal or impaired glucose tolerance. Thus the term "glucolipotoxicity" is perhaps more appropriate in describing the phenomenon. Several mechanisms have been proposed to explain glucolipotoxicity-induced β-cell dysfunction and death, but its major factors appear to be depression of key transcription factor gene expression by altered intracellular energy metabolism and oxidative stress. Therefore, stabilization of metabolic changes induced by glucolipotoxicity in β-cells represents a new avenue for the treatment of type 2 diabetes mellitus.

Citations

Citations to this article as recorded by  
  • Intravoxel incoherent motion diffusion-weighted imaging of pancreas: Probing evidence of β-cell dysfunction in asymptomatic adults with hyperglycemia in vivo
    Yingying Song, Bo Chen, Kejing Zeng, Kejia Cai, Hui Sun, Deqing Liu, Ping Liu, Gugen Xu, Guihua Jiang
    Magnetic Resonance Imaging.2024; 108: 161.     CrossRef
  • PIMT Controls Insulin Synthesis and Secretion through PDX1
    Rahul Sharma, Sujay K. Maity, Partha Chakrabarti, Madhumohan R. Katika, Satyamoorthy Kapettu, Kishore V. L. Parsa, Parimal Misra
    International Journal of Molecular Sciences.2023; 24(9): 8084.     CrossRef
  • Growth differentiation factor-15 prevents glucotoxicity and connexin-36 downregulation in pancreatic beta-cells
    Mohamed Asrih, Rodolphe Dusaulcy, Yvan Gosmain, Jacques Philippe, Emmanuel Somm, François R. Jornayvaz, Baeki E. Kang, Yunju Jo, Min Jeong Choi, Hyon-Seung Yi, Dongryeol Ryu, Karim Gariani
    Molecular and Cellular Endocrinology.2022; 541: 111503.     CrossRef
  • Oxidative stress in the pathophysiology of type 2 diabetes and related complications: Current therapeutics strategies and future perspectives
    Jasvinder Singh Bhatti, Abhishek Sehrawat, Jayapriya Mishra, Inderpal Singh Sidhu, Umashanker Navik, Naina Khullar, Shashank Kumar, Gurjit Kaur Bhatti, P. Hemachandra Reddy
    Free Radical Biology and Medicine.2022; 184: 114.     CrossRef
  • Phenotypic Screening for Small Molecules that Protect β-Cells from Glucolipotoxicity
    Jonnell C. Small, Aidan Joblin-Mills, Kaycee Carbone, Maria Kost-Alimova, Kumiko Ayukawa, Carol Khodier, Vlado Dancik, Paul A. Clemons, Andrew B. Munkacsi, Bridget K. Wagner
    ACS Chemical Biology.2022; 17(5): 1131.     CrossRef
  • Oxidative stress-mediated beta cell death and dysfunction as a target for diabetes management
    Svetlana Dinić, Jelena Arambašić Jovanović, Aleksandra Uskoković, Mirjana Mihailović, Nevena Grdović, Anja Tolić, Jovana Rajić, Marija Đorđević, Melita Vidaković
    Frontiers in Endocrinology.2022;[Epub]     CrossRef
  • Targeting pancreatic β cells for diabetes treatment
    Chirag Jain, Ansarullah, Sara Bilekova, Heiko Lickert
    Nature Metabolism.2022; 4(9): 1097.     CrossRef
  • The high-fructose intake of dams during pregnancy and lactation exerts sex-specific effects on adult rat offspring metabolism
    Francisca A. Tobar-Bernal, Sergio R. Zamudio, Lucía Quevedo-Corona
    Journal of Developmental Origins of Health and Disease.2021; 12(3): 411.     CrossRef
  • Mechanisms of Beta-Cell Apoptosis in Type 2 Diabetes-Prone Situations and Potential Protection by GLP-1-Based Therapies
    Safia Costes, Gyslaine Bertrand, Magalie A. Ravier
    International Journal of Molecular Sciences.2021; 22(10): 5303.     CrossRef
  • A Review of Current Trends with Type 2 Diabetes Epidemiology, Aetiology, Pathogenesis, Treatments and Future Perspectives
    Josh Reed, Stephen Bain, Venkateswarlu Kanamarlapudi
    Diabetes, Metabolic Syndrome and Obesity: Targets and Therapy.2021; Volume 14: 3567.     CrossRef
  • Beta-Cell Dysfunction Induced by Tacrolimus: A Way to Explain Type 2 Diabetes?
    Ana Elena Rodriguez-Rodriguez, Esteban Porrini, Armando Torres
    International Journal of Molecular Sciences.2021; 22(19): 10311.     CrossRef
  • Involvement of Metabolic Lipid Mediators in the Regulation of Apoptosis
    Piotr Wójcik, Neven Žarković, Agnieszka Gęgotek, Elżbieta Skrzydlewska
    Biomolecules.2020; 10(3): 402.     CrossRef
  • Phytochemicals as modulators of β-cells and immunity for the therapy of type 1 diabetes: Recent discoveries in pharmacological mechanisms and clinical potential
    Maria Karmella Apaya, Tien-Fen Kuo, Meng-Ting Yang, Greta Yang, Chiao-Ling Hsiao, Song-Bin Chang, Yenshou Lin, Wen-Chin Yang
    Pharmacological Research.2020; 156: 104754.     CrossRef
  • Clinical implications, diagnosis, and management of diabetes in patients with chronic liver diseases
    Waihong Chung, Kittichai Promrat, Jack Wands
    World Journal of Hepatology.2020; 12(9): 533.     CrossRef
  • Mitochondrial GTP Links Nutrient Sensing to β Cell Health, Mitochondrial Morphology, and Insulin Secretion Independent of OxPhos
    Sean R. Jesinkey, Anila K. Madiraju, Tiago C. Alves, OrLando H. Yarborough, Rebecca L. Cardone, Xiaojian Zhao, Yassmin Parsaei, Ali R. Nasiri, Gina Butrico, Xinran Liu, Anthony J. Molina, Austin M. Rountree, Adam S. Neal, Dane M. Wolf, John Sterpka, Willi
    Cell Reports.2019; 28(3): 759.     CrossRef
  • Metabolic signatures suggest o-phosphocholine to UDP-N-acetylglucosamine ratio as a potential biomarker for high-glucose and/or palmitate exposure in pancreatic β-cells
    Saleem Yousf, Devika M. Sardesai, Abraham B. Mathew, Rashi Khandelwal, Jhankar D. Acharya, Shilpy Sharma, Jeetender Chugh
    Metabolomics.2019;[Epub]     CrossRef
  • Glucolipotoxicity Alters Insulin Secretion via Epigenetic Changes in Human Islets
    Elin Hall, Josefine Jönsson, Jones K. Ofori, Petr Volkov, Alexander Perfilyev, Marloes Dekker Nitert, Lena Eliasson, Charlotte Ling, Karl Bacos
    Diabetes.2019; 68(10): 1965.     CrossRef
  • Bioinformatic analysis of peripheral blood RNA-sequencing sensitively detects the cause of late graft loss following overt hyperglycemia in pig-to-nonhuman primate islet xenotransplantation
    Hyun-Je Kim, Ji Hwan Moon, Hyunwoo Chung, Jun-Seop Shin, Bongi Kim, Jong-Min Kim, Jung-Sik Kim, Il-Hee Yoon, Byoung-Hoon Min, Seong-Jun Kang, Yong-Hee Kim, Kyuri Jo, Joungmin Choi, Heejoon Chae, Won-Woo Lee, Sun Kim, Chung-Gyu Park
    Scientific Reports.2019;[Epub]     CrossRef
  • Centaurium erythraea extract improves survival and functionality of pancreatic beta-cells in diabetes through multiple routes of action
    Miloš Đorđević, Nevena Grdović, Mirjana Mihailović, Jelena Arambašić Jovanović, Aleksandra Uskoković, Jovana Rajić, Marija Sinadinović, Anja Tolić, Danijela Mišić, Branislav Šiler, Goran Poznanović, Melita Vidaković, Svetlana Dinić
    Journal of Ethnopharmacology.2019; 242: 112043.     CrossRef
  • Caveolin-1 deficiency protects pancreatic β cells against palmitate-induced dysfunction and apoptosis
    Wen Zeng, Jiansong Tang, Haicheng Li, Haixia Xu, Hongyun Lu, Hangya Peng, Chuwen Lin, Rili Gao, Shuo Lin, Keyi Lin, Kunying Liu, Yan Jiang, Jianping Weng, Longyi Zeng
    Cellular Signalling.2018; 47: 65.     CrossRef
  • Temporal Proteomic Analysis of Pancreatic β-Cells in Response to Lipotoxicity and Glucolipotoxicity
    Zonghong Li, Hongyang Liu, Zhangjing Niu, Wen Zhong, Miaomiao Xue, Jifeng Wang, Fuquan Yang, Yue Zhou, Yifa Zhou, Tao Xu, Junjie Hou
    Molecular & Cellular Proteomics.2018; 17(11): 2119.     CrossRef
  • Weight-Independent Mechanisms of Glucose Control After Roux-en-Y Gastric Bypass
    Blandine Laferrère, François Pattou
    Frontiers in Endocrinology.2018;[Epub]     CrossRef
  • Effects of moderate exercise on biochemical, morphological, and physiological parameters of the pancreas of female mice with estrogen deprivation and dyslipidemia
    Aparecida Gabriela Bexiga Veloso, Nathalia Edviges Alves Lima, Elisabete de Marco Ornelas, Clever Gomes Cardoso, Mara Rubia Marques, Beatriz da Costa Aguiar Alves Reis, Fernando Luiz Affonso Fonseca, Laura Beatriz Mesiano Maifrino
    Medical Molecular Morphology.2018; 51(2): 118.     CrossRef
  • Cardiac Glucolipotoxicity and Cardiovascular Outcomes
    Marlon E. Cerf
    Medicina.2018; 54(5): 70.     CrossRef
  • High frequency of type 2 diabetes and impaired glucose tolerance in Japanese subjects with the angiopoietin-like protein 8 R59W variant
    Jianhui Liu, Kunimasa Yagi, Atsushi Nohara, Daisuke Chujo, Azusa Ohbatake, Aya Fujimoto, Yukiko Miyamoto, Junji Kobayashi, Masakazu Yamagishi
    Journal of Clinical Lipidology.2018; 12(2): 331.     CrossRef
  • Identification of Digestive Enzyme Inhibitors from Ludwigia octovalvis (Jacq.) P.H.Raven
    Dulce Morales, Guillermo Ramirez, Armando Herrera-Arellano, Jaime Tortoriello, Miguel Zavala, Alejandro Zamilpa
    Evidence-Based Complementary and Alternative Medicine.2018; 2018: 1.     CrossRef
  • The impairment of glucose-stimulated insulin secretion in pancreatic β-cells caused by prolonged glucotoxicity and lipotoxicity is associated with elevated adaptive antioxidant response
    Jingqi Fu, Qi Cui, Bei Yang, Yongyong Hou, Huihui Wang, Yuanyuan Xu, Difei Wang, Qiang Zhang, Jingbo Pi
    Food and Chemical Toxicology.2017; 100: 161.     CrossRef
  • Phenolic constituents and modulatory effects of Raffia palm leaf ( Raphia hookeri ) extract on carbohydrate hydrolyzing enzymes linked to type-2 diabetes
    Felix A. Dada, Sunday I. Oyeleye, Opeyemi B. Ogunsuyi, Tosin A. Olasehinde, Stephen A. Adefegha, Ganiyu Oboh, Aline A. Boligon
    Journal of Traditional and Complementary Medicine.2017; 7(4): 494.     CrossRef
  • A gene-environment interaction analysis of plasma selenium with prevalent and incident diabetes: The Hortega study
    Inmaculada Galan-Chilet, Maria Grau-Perez, Griselda De Marco, Eliseo Guallar, Juan Carlos Martin-Escudero, Alejandro Dominguez-Lucas, Isabel Gonzalez-Manzano, Raul Lopez-Izquierdo, Laisa Socorro Briongos-Figuero, Josep Redon, Felipe Javier Chaves, Maria T
    Redox Biology.2017; 12: 798.     CrossRef
  • Differential dose-dependent effects of zinc oxide nanoparticles on oxidative stress-mediated pancreatic β-cell death
    Swati C Asani, Rinku D Umrani, Kishore M Paknikar
    Nanomedicine.2017; 12(7): 745.     CrossRef
  • Mild hyperglycemia triggered islet function recovery in streptozotocin‐induced insulin‐deficient diabetic rats
    Yu Cheng, Jing Shen, Weizheng Ren, Haojie Hao, Zongyan Xie, Jiejie Liu, Yiming Mu, Weidong Han
    Journal of Diabetes Investigation.2017; 8(1): 44.     CrossRef
  • The Novel Mechanisms Concerning the Inhibitions of Palmitate-Induced Proinflammatory Factor Releases and Endogenous Cellular Stress with Astaxanthin on MIN6 β-Cells
    Atsuko Kitahara, Kazuto Takahashi, Naru Morita, Toshitaka Murashima, Hirohisa Onuma, Yoshikazu Sumitani, Toshiaki Tanaka, Takuma Kondo, Toshio Hosaka, Hitoshi Ishida
    Marine Drugs.2017; 15(6): 185.     CrossRef
  • Protein kinase STK25 aggravates the severity of non-alcoholic fatty pancreas disease in mice
    Esther Nuñez-Durán, Belén Chanclón, Silva Sütt, Joana Real, Hanns-Ulrich Marschall, Ingrid Wernstedt Asterholm, Emmelie Cansby, Margit Mahlapuu
    Journal of Endocrinology.2017; 234(1): 15.     CrossRef
  • Metformin prevents glucotoxicity by alleviating oxidative and ER stress–induced CD36 expression in pancreatic beta cells
    Jun Sung Moon, Udayakumar Karunakaran, Suma Elumalai, In-Kyu Lee, Hyoung Woo Lee, Yong-Woon Kim, Kyu Chang Won
    Journal of Diabetes and its Complications.2017; 31(1): 21.     CrossRef
  • The Glucotoxicity Protecting Effect of Ezetimibe in Pancreatic Beta Cells via Inhibition of CD36
    Ji Sung Yoon, Jun Sung Moon, Yong-Woon Kim, Kyu Chang Won, Hyoung Woo Lee
    Journal of Korean Medical Science.2016; 31(4): 547.     CrossRef
  • The level of netrin-1 is decreased in newly diagnosed type 2 diabetes mellitus patients
    Chenxiao Liu, Xianjin Ke, Ying Wang, Xiu Feng, Qi Li, Ying Zhang, Jian Zhu, Qian Li
    BMC Endocrine Disorders.2016;[Epub]     CrossRef
  • Liraglutide Compromises Pancreatic β Cell Function in a Humanized Mouse Model
    Midhat H. Abdulreda, Rayner Rodriguez-Diaz, Alejandro Caicedo, Per-Olof Berggren
    Cell Metabolism.2016; 23(3): 541.     CrossRef
  • Glucolipotoxicity diminishes cardiomyocyte TFEB and inhibits lysosomal autophagy during obesity and diabetes
    Purvi C. Trivedi, Jordan J. Bartlett, Lester J. Perez, Keith R. Brunt, Jean Francois Legare, Ansar Hassan, Petra C. Kienesberger, Thomas Pulinilkunnil
    Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids.2016; 1861(12): 1893.     CrossRef
  • Diabetes risk prediction model for non‐obese Asian Indians residing in North India using cut‐off values for pancreatic and intra‐abdominal fat volume and liver span
    Shajith Anoop, Anoop Misra, Kalaivani Mani, Ravindra Mohan Pandey, Seema Gulati
    Journal of Diabetes.2016; 8(5): 729.     CrossRef
  • Identifying crop variants with high resistant starch content to maintain healthy glucose homeostasis
    K. Petropoulou, E. S. Chambers, D. J. Morrison, T. Preston, I. F. Godsland, P. Wilde, A. Narbad, R. Parker, L. Salt, V. J. Morris, C. Domoney, S. J. Persaud, E. Holmes, S. Penson, J. Watson, M. Stocks, M. Buurman, M. Luterbacher, G. Frost
    Nutrition Bulletin.2016; 41(4): 372.     CrossRef
  • Endogenous GLP-1 as a key self-defense molecule against lipotoxicity in pancreatic islets
    CHENGHU HUANG, LI YUAN, SHUYI CAO
    International Journal of Molecular Medicine.2015; 36(1): 173.     CrossRef
  • Mitochondrien als Kraftwerk der β-Zelle
    S. Baltrusch, F. Reinhardt, M. Tiedge
    Der Diabetologe.2015; 11(3): 231.     CrossRef
  • Protein-Binding Function of RNA-Dependent Protein Kinase Promotes Proliferation through TRAF2/RIP1/NF-κB/c-Myc Pathway in Pancreatic β cells
    LiLi Gao, Wei Tang, ZhengZheng Ding, DingYu Wang, XiaoQiang Qi, HuiWen Wu, Jun Guo
    Molecular Medicine.2015; 21(1): 154.     CrossRef
  • Phosphorylation of caveolin-1 on tyrosine-14 induced by ROS enhances palmitate-induced death of beta-pancreatic cells
    Sergio Wehinger, Rina Ortiz, María Inés Díaz, Adam Aguirre, Manuel Valenzuela, Paola Llanos, Christopher Mc Master, Lisette Leyton, Andrew F.G. Quest
    Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease.2015; 1852(5): 693.     CrossRef
  • Body Fat Patterning, Hepatic Fat and Pancreatic Volume of Non-Obese Asian Indians with Type 2 Diabetes in North India: A Case-Control Study
    Anoop Misra, Shajith Anoop, Seema Gulati, Kalaivani Mani, Surya Prakash Bhatt, Ravindra Mohan Pandey, Anna Alisi
    PLOS ONE.2015; 10(10): e0140447.     CrossRef
  • CD36 initiated signaling mediates ceramide-induced TXNIP expression in pancreatic beta-cells
    Udayakumar Karunakaran, Jun Sung Moon, Hyoung Woo Lee, Kyu Chang Won
    Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease.2015; 1852(11): 2414.     CrossRef
  • Acarbose, lente carbohydrate, and prebiotics promote metabolic health and longevity by stimulating intestinal production of GLP-1
    Mark F McCarty, James J DiNicolantonio
    Open Heart.2015; 2(1): e000205.     CrossRef
  • P38 Plays an Important Role in Glucolipotoxicity-Induced Apoptosis in INS-1 Cells
    Lingli Zhou, Xiaoling Cai, Xueyao Han, Linong Ji
    Journal of Diabetes Research.2014; 2014: 1.     CrossRef
  • ImpairedβCell Function in Chinese Newly Diagnosed Type 2 Diabetes Mellitus with Hyperlipidemia
    Yuhang Ma, Yufan Wang, Qianfang Huang, Qian Ren, Su Chen, Aifang Zhang, Li Zhao, Qin Zhen, Yongde Peng
    Journal of Diabetes Research.2014; 2014: 1.     CrossRef
  • Type 2 Diabetes, PUFAs, and Vitamin D: Their Relation to Inflammation
    Ana L. Guadarrama-López, Roxana Valdés-Ramos, Beatríz E. Martínez-Carrillo
    Journal of Immunology Research.2014; 2014: 1.     CrossRef
  • A Comparative Study of the Effects of a Dipeptidyl Peptidase-IV Inhibitor and Sulfonylurea on Glucose Variability in Patients with Type 2 Diabetes with Inadequate Glycemic Control on Metformin
    Hun-Sung Kim, Jeong-Ah Shin, Seung-Hwan Lee, Eun-Sook Kim, Jae-Hyoung Cho, Ho-Young Son, Kun-Ho Yoon
    Diabetes Technology & Therapeutics.2013; 15(10): 810.     CrossRef
  • Localization and regulation of pancreatic selenoprotein P
    Holger Steinbrenner, Anna-Lena Hotze, Bodo Speckmann, Antonio Pinto, Helmut Sies, Matthias Schott, Margret Ehlers, Werner A Scherbaum, Sven Schinner
    Journal of Molecular Endocrinology.2013; 50(1): 31.     CrossRef
  • Glycated Albumin Causes Pancreatic β-Cells Dysfunction Through Autophagy Dysfunction
    Young Mi Song, Sun Ok Song, Young-Hye You, Kun-Ho Yoon, Eun Seok Kang, Bong Soo Cha, Hyun Chul Lee, Ji-Won Kim, Byung-Wan Lee
    Endocrinology.2013; 154(8): 2626.     CrossRef
  • miRNA-30a-5p-mediated silencing of Beta2/NeuroD expression is an important initial event of glucotoxicity-induced beta cell dysfunction in rodent models
    J.-W. Kim, Y.-H. You, S. Jung, H. Suh-Kim, I.-K. Lee, J.-H. Cho, K.-H. Yoon
    Diabetologia.2013; 56(4): 847.     CrossRef
  • Investigating Endothelial Activation and Oxidative Stress in relation to Glycaemic Control in a Multiethnic Population
    E. M. Brady, D. R. Webb, D. H. Morris, K. Khunti, D. S. C. Talbot, N. Sattar, M. J. Davies
    Experimental Diabetes Research.2012; 2012: 1.     CrossRef
  • Inhibition of fatty acid translocase cluster determinant 36 (CD36), stimulated by hyperglycemia, prevents glucotoxicity in INS-1 cells
    Yong-Woon Kim, Jun Sung Moon, Ye Jin Seo, So-Young Park, Jong-Yeon Kim, Ji Sung Yoon, In-Kyu Lee, Hyoung Woo Lee, Kyu Chang Won
    Biochemical and Biophysical Research Communications.2012; 420(2): 462.     CrossRef
  • Human Monoclonal Antibodies against Glucagon Receptor Improve Glucose Homeostasis by Suppression of Hepatic Glucose Output in Diet-Induced Obese Mice
    Wook-Dong Kim, Yong-ho Lee, Min-Hee Kim, Sun-Young Jung, Woo-Chan Son, Seon-Joo Yoon, Byung-Wan Lee, Nigel Irwin
    PLoS ONE.2012; 7(12): e50954.     CrossRef
  • Understanding Pancreas Development for β-Cell Repair and Replacement Therapies
    Aurelia Raducanu, Heiko Lickert
    Current Diabetes Reports.2012; 12(5): 481.     CrossRef

Diabetes Metab J : Diabetes & Metabolism Journal