Skip Navigation
Skip to contents

Diabetes Metab J : Diabetes & Metabolism Journal

Search
OPEN ACCESS

Search

Page Path
HOME > Search
6 "Ji-Won Kim"
Filter
Filter
Article category
Keywords
Publication year
Authors
Funded articles
Original Articles
Basic Research
Differentiation of Microencapsulated Neonatal Porcine Pancreatic Cell Clusters in Vitro Improves Transplant Efficacy in Type 1 Diabetes Mellitus Mice
Gyeong-Jin Cheon, Heon-Seok Park, Eun-Young Lee, Min Jung Kim, Young-Hye You, Marie Rhee, Ji-Won Kim, Kun-Ho Yoon
Diabetes Metab J. 2022;46(5):677-688.   Published online February 7, 2022
DOI: https://doi.org/10.4093/dmj.2021.0202
  • 4,521 View
  • 252 Download
  • 2 Web of Science
  • 2 Crossref
AbstractAbstract PDFSupplementary MaterialPubReader   ePub   
Background
Neonatal porcine pancreatic cell clusters (NPCCs) have been proposed as an alternative source of β cells for islet transplantation because of their low cost and growth potential after transplantation. However, the delayed glucose lowering effect due to the immaturity of NPCCs and immunologic rejection remain as a barrier to NPCC’s clinical application. Here, we demonstrate accelerated differentiation and immune-tolerant NPCCs by in vitro chemical treatment and microencapsulation.
Methods
NPCCs isolated from 3-day-old piglets were cultured in F-10 media and then microencapsulated with alginate on day 5. Differentiation of NPCCs is facilitated by media supplemented with activin receptor-like kinase 5 inhibitor II, triiodothyronine and exendin-4 for 2 weeks. Marginal number of microencapsulated NPCCs to cure diabetes with and without differentiation were transplanted into diabetic mice and observed for 8 weeks.
Results
The proportion of insulin-positive cells and insulin mRNA levels of NPCCs were significantly increased in vitro in the differentiated group compared with the undifferentiated group. Blood glucose levels decreased eventually after transplantation of microencapsulated NPCCs in diabetic mice and normalized after 7 weeks in the differentiated group. In addition, the differentiated group showed nearly normal glucose tolerance at 8 weeks after transplantation. In contrast, neither blood glucose levels nor glucose tolerance were improved in the undifferentiated group. Retrieved graft in the differentiated group showed greater insulin response to high glucose compared with the undifferentiated group.
Conclusion
in vitro differentiation of microencapsulated immature NPCCs increased the proportion of insulin-positive cells and improved transplant efficacy in diabetic mice without immune rejection.

Citations

Citations to this article as recorded by  
  • Dual-targeted nano-encapsulation of neonatal porcine islet-like cell clusters with triiodothyronine-loaded bifunctional polymersomes
    Sang Hoon Lee, Minse Kim, Eun-Jin Lee, Sun Mi Ahn, Yu-Rim Ahn, Jaewon Choi, Jung-Taek Kang, Hyun-Ouk Kim
    Discover Nano.2024;[Epub]     CrossRef
  • Long‐term efficacy of encapsulated xenogeneic islet transplantation: Impact of encapsulation techniques and donor genetic traits
    Heon‐Seok Park, Eun Young Lee, Young‐Hye You, Marie Rhee, Jong‐Min Kim, Seong‐Soo Hwang, Poong‐Yeon Lee
    Journal of Diabetes Investigation.2024;[Epub]     CrossRef
Others
Generation of Insulin-Expressing Cells in Mouse Small Intestine by Pdx1, MafA, and BETA2/NeuroD
So-Hyun Lee, Marie Rhee, Ji-Won Kim, Kun-Ho Yoon
Diabetes Metab J. 2017;41(5):405-416.   Published online September 5, 2017
DOI: https://doi.org/10.4093/dmj.2017.41.5.405
  • 5,106 View
  • 64 Download
  • 5 Web of Science
  • 5 Crossref
AbstractAbstract PDFSupplementary MaterialPubReader   
Background

To develop surrogate insulin-producing cells for diabetes therapy, adult stem cells have been identified in various tissues and studied for their conversion into β-cells. Pancreatic progenitor cells are derived from the endodermal epithelium and formed in a manner similar to gut progenitor cells. Here, we generated insulin-producing cells from the intestinal epithelial cells that induced many of the specific pancreatic transcription factors using adenoviral vectors carrying three genes: PMB (pancreatic and duodenal homeobox 1 [Pdx1], V-maf musculoaponeurotic fibrosarcoma oncogene homolog A [MafA], and BETA2/NeuroD).

Methods

By direct injection into the intestine through the cranial mesenteric artery, adenoviruses (Ad) were successfully delivered to the entire intestine. After virus injection, we could confirm that the small intestine of the mouse was appropriately infected with the Ad-Pdx1 and triple Ad-PMB.

Results

Four weeks after the injection, insulin mRNA was expressed in the small intestine, and the insulin gene expression was induced in Ad-Pdx1 and Ad-PMB compared to control Ad-green fluorescent protein. In addition, the conversion of intestinal cells into insulin-expressing cells was detected in parts of the crypts and villi located in the small intestine.

Conclusion

These data indicated that PMB facilitate the differentiation of mouse intestinal cells into insulin-expressing cells. In conclusion, the small intestine is an accessible and abundant source of surrogate insulin-producing cells.

Citations

Citations to this article as recorded by  
  • Harnessing gut cells for functional insulin production: Strategies and challenges
    Kelvin Baafi, John C. March
    Biotechnology Notes.2023; 4: 7.     CrossRef
  • Differential Morphological Diagnosis of Various Forms of Congenital Hyperinsulinism in Children
    Lubov Borisovna Mitrofanova, Anastasia Arkadyevna Perminova, Daria Viktorovna Ryzhkova, Anna Andreyevna Sukhotskaya, Vladimir Gireyevich Bairov, Irina Leorovna Nikitina
    Frontiers in Endocrinology.2021;[Epub]     CrossRef
  • Generation of iPSC-derived insulin-producing cells from patients with type 1 and type 2 diabetes compared with healthy control
    Min Jung Kim, Eun Young Lee, Young-Hye You, Hae Kyung Yang, Kun-Ho Yoon, Ji-Won Kim
    Stem Cell Research.2020; 48: 101958.     CrossRef
  • ERK Regulates NeuroD1-mediated Neurite Outgrowth via Proteasomal Degradation
    Tae-young Lee, In-Su Cho, Narayan Bashyal, Francisco J Naya, Ming-Jer Tsai, Jeong Seon Yoon, Jung-Mi Choi, Chang-Hwan Park, Sung-Soo Kim, Haeyoung Suh-Kim
    Experimental Neurobiology.2020; 29(3): 189.     CrossRef
  • Generation of a PDX1–EGFP reporter human induced pluripotent stem cell line, KSCBi005-A-3, using the CRISPR/Cas9 system
    Youngsun Lee, Hye Young Choi, Ara Kwon, Hyeyeon Park, Mi-Hyun Park, Ji-Won Kim, Min Jung Kim, Yong-Ou Kim, Sungwook Kwak, Soo Kyung Koo
    Stem Cell Research.2019; 41: 101632.     CrossRef
Pattern of Stress-Induced Hyperglycemia according to Type of Diabetes: A Predator Stress Model
Jin-Sun Chang, Young-Hye You, Shin-Young Park, Ji-Won Kim, Hun-Sung Kim, Kun-Ho Yoon, Jae-Hyoung Cho
Diabetes Metab J. 2013;37(6):475-483.   Published online December 12, 2013
DOI: https://doi.org/10.4093/dmj.2013.37.6.475
  • 4,050 View
  • 50 Download
  • 5 Crossref
AbstractAbstract PDFPubReader   
Background

We aimed to quantify stress-induced hyperglycemia and differentiate the glucose response between normal animals and those with diabetes. We also examined the pattern in glucose fluctuation induced by stress according to type of diabetes.

Methods

To load psychological stress on animal models, we used a predator stress model by exposing rats to a cat for 60 minutes and measured glucose level from the beginning to the end of the test to monitor glucose fluctuation. We induced type 1 diabetes model (T1D) for ten Sprague-Dawley rats using streptozotocin and used five Otsuka Long-Evans Tokushima Fatty rats as obese type 2 diabetes model (OT2D) and 10 Goto-Kakizaki rats as nonobese type 2 diabetes model (NOT2D). We performed the stress loading test in both the normal and diabetic states and compared patterns of glucose fluctuation among the three models. We classified the pattern of glucose fluctuation into A, B, and C types according to speed of change in glucose level.

Results

Increase in glucose, total amount of hyperglycemic exposure, time of stress-induced hyperglycemia, and speed of glucose increase were significantly increased in all models compared to the normal state. While the early increase in glucose after exposure to stress was higher in T1D and NOT2D, it was slower in OT2D. The rate of speed of the decrease in glucose level was highest in NOT2D and lowest in OT2D.

Conclusion

The diabetic state was more vulnerable to stress compared to the normal state in all models, and the pattern of glucose fluctuation differed among the three types of diabetes. The study provides basic evidence for stress-induced hyperglycemia patterns and characteristics used for the management of diabetes patients.

Citations

Citations to this article as recorded by  
  • Stress hyperglycemia as first sign of asymptomatic type 1 diabetes: an instructive case
    Wei-De Wang, Chun-Hao Chu, Chiung-Hsi Tien, Shuo-Yu Wang, Shih-Yao Liu, Chien-Ming Lin
    BMC Pediatrics.2021;[Epub]     CrossRef
  • Genetic determinants of obesity heterogeneity in type II diabetes
    Somayeh Alsadat Hosseini Khorami, Mohd Sokhini Abd Mutalib, Mohammad Feili Shiraz, Joseph Anthony Abdullah, Zulida Rejali, Razana Mohd Ali, Huzwah Khaza’ai
    Nutrition & Metabolism.2020;[Epub]     CrossRef
  • Sex Dimorphic Responses of the Hypothalamus–Pituitary–Thyroid Axis to Maternal Separation and Palatable Diet
    Lorraine Jaimes-Hoy, Fidelia Romero, Jean-Louis Charli, Patricia Joseph-Bravo
    Frontiers in Endocrinology.2019;[Epub]     CrossRef
  • Hesperidin protects against stress induced gastric ulcer through regulation of peroxisome proliferator activator receptor gamma in diabetic rats
    Shimaa M. Elshazly, Dalia M. Abd El Motteleb, Islam A.A.E-H. Ibrahim
    Chemico-Biological Interactions.2018; 291: 153.     CrossRef
  • Physiology and Neurobiology of Stress and the Implications for Physical Health
    B Sivaprakash
    Annals of SBV.2014; 3(1): 25.     CrossRef
Review
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
  • 54,436 View
  • 89 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
Original Articles
Adenoviruses Expressing PDX-1, BETA2/NeuroD and MafA Induces the Transdifferentiation of Porcine Neonatal Pancreas Cell Clusters and Adult Pig Pancreatic Cells into Beta-Cells
Young-Hye You, Dong-Sik Ham, Heon-Seok Park, Marie Rhee, Ji-Won Kim, Kun-Ho Yoon
Diabetes Metab J. 2011;35(2):119-129.   Published online April 30, 2011
DOI: https://doi.org/10.4093/dmj.2011.35.2.119
  • 3,472 View
  • 36 Download
  • 9 Crossref
AbstractAbstract PDFPubReader   
Background

A limitation in the number of insulin-producing pancreatic beta-cells is a special feature of diabetes. The identification of alternative sources for the induction of insulin-producing surrogate beta-cells is a matter of profound importance. PDX-1/VP16, BETA2/NeuroD, and MafA overexpression have been shown to influence the differentiation and proliferation of pancreatic stem cells. However, few studies have been conducted using adult animal pancreatic stem cells.

Methods

Adult pig pancreatic cells were prepared from the non-endocrine fraction of adult pig pancreata. Porcine neonatal pancreas cell clusters (NPCCs) were prepared from neonatal pigs aged 1-2 days. The dispersed pancreatic cells were infected with PDX-1/VP16, BETA2/NeuroD, and MafA adenoviruses. After infection, these cells were transplanted under the kidney capsules of normoglycemic nude mice.

Results

The adenovirus-mediated overexpression of PDX-1, BETA2/NeuroD and MafA induced insulin gene expression in NPCCs, but not in adult pig pancreatic cells. Immunocytochemistry revealed that the number of insulin-positive cells in NPCCs and adult pig pancreatic cells was approximately 2.6- and 1.1-fold greater than those in the green fluorescent protein control group, respectively. At four weeks after transplantation, the relative volume of insulin-positive cells in the grafts increased in the NPCCs, but not in the adult porcine pancreatic cells.

Conclusion

These data indicate that PDX-1, BETA2/NeuroD, and MafA facilitate the beta-cell differentiation of NPCCs, but not adult pig pancreatic cells. Therefore PDX-1, BETA2/NeuroD, and MafA-induced NPCCs can be considered good sources for the induction of pancreatic beta-cells, and may also have some utility in the treatment of diabetes.

Citations

Citations to this article as recorded by  
  • The pig pangenome provides insights into the roles of coding structural variations in genetic diversity and adaptation
    Zhengcao Li, Xiaohong Liu, Chen Wang, Zhenyang Li, Bo Jiang, Ruifeng Zhang, Lu Tong, Youping Qu, Sheng He, Haifan Chen, Yafei Mao, Qingnan Li, Torsten Pook, Yu Wu, Yanjun Zan, Hui Zhang, Lu Li, Keying Wen, Yaosheng Chen
    Genome Research.2023; 33(10): 1833.     CrossRef
  • Improvement of the therapeutic capacity of insulin-producing cells trans-differentiated from human liver cells using engineered cell sheet
    Yu Na Lee, Hye-Jin Yi, Eun Hye Seo, Jooyun Oh, Song Lee, Sarah Ferber, Teruo Okano, In Kyong Shim, Song Cheol Kim
    Stem Cell Research & Therapy.2021;[Epub]     CrossRef
  • Generation of iPSC-derived insulin-producing cells from patients with type 1 and type 2 diabetes compared with healthy control
    Min Jung Kim, Eun Young Lee, Young-Hye You, Hae Kyung Yang, Kun-Ho Yoon, Ji-Won Kim
    Stem Cell Research.2020; 48: 101958.     CrossRef
  • Effect of FIGF overexpression on liver cells transforming to insulin-producing cells
    Yaqin He, Xiaoliang Xie, Xiaoyan Li, Shikuo Rong, Yukui Li, Zhenhui Lu
    Journal of Biosciences.2019;[Epub]     CrossRef
  • Generation of Insulin-Expressing Cells in Mouse Small Intestine by Pdx1, MafA, and BETA2/NeuroD
    So-Hyun Lee, Marie Rhee, Ji-Won Kim, Kun-Ho Yoon
    Diabetes & Metabolism Journal.2017; 41(5): 405.     CrossRef
  • Quantitative Raman spectral changes of the differentiation of mesenchymal stem cells into islet-like cells by biochemical component analysis and multiple peak fitting
    Xin Su, Shaoyin Fang, Daosen Zhang, Qinnan Zhang, Yingtian He, Xiaoxu Lu, Shengde Liu, Liyun Zhong
    Journal of Biomedical Optics.2015; 20(12): 125002.     CrossRef
  • Generation of Functional Insulin-Producing Cells from Neonatal Porcine Liver-Derived Cells by PDX1/VP16, BETA2/NeuroD and MafA
    Dong-Sik Ham, Juyoung Shin, Ji-Won Kim, Heon-Seok Park, Jae-Hyoung Cho, Kun-Ho Yoon, Kathrin Maedler
    PLoS ONE.2013; 8(11): e79076.     CrossRef
  • PPARγ Activation Attenuates Glycated-Serum Induced Pancreatic Beta-Cell Dysfunction through Enhancing Pdx1 and Mafa Protein Stability
    Yunxia Zhu, Ai Ma, Hongxiu Zhang, Chaojun Li, Rebecca Berdeaux
    PLoS ONE.2013; 8(2): e56386.     CrossRef
  • β‐Cell differentiation and regeneration in type 1 diabetes
    L. Ding, C. Gysemans, C. Mathieu
    Diabetes, Obesity and Metabolism.2013; 15(s3): 98.     CrossRef
Repeated Gene Transfection Impairs the Engraftment of Transplanted Porcine Neonatal Pancreatic Cells
Min Koo Seo, Cheng-Lin Sun, Ji-Won Kim, Kun-Ho Yoon, Suk Kyeong Lee
Diabetes Metab J. 2011;35(1):72-79.   Published online February 28, 2011
DOI: https://doi.org/10.4093/dmj.2011.35.1.72
  • 28,573 View
  • 27 Download
  • 3 Crossref
AbstractAbstract PDFPubReader   
Background

Previously, we reported that neonatal porcine pancreatic cells transfected with hepatocyte growth factor (HGF) gene in an Epstein-Barr virus (EBV)-based plasmid (pEBVHGF) showed improved proliferation and differentiation compared to those of the control. In this study, we examined if pancreatic cells transfected repeatedly with pEBVHGF can be successfully grafted to control blood glucose in a diabetes mouse model.

Methods

Neonatal porcine pancreatic cells were cultured as a monolayer and were transfected with pEBVHGF every other day for a total of three transfections. The transfected pancreatic cells were re-aggregated and transplanted into kidney capsules of diabetic nude mice or normal nude mice. Blood glucose level and body weight were measured every other day after transplantation. The engraftment of the transplanted cells and differentiation into beta cells were assessed using immunohistochemistry.

Results

Re-aggregation of the pancreatic cells before transplantation improved engraftment of the cells and facilitated neovascularization of the graft. Right before transplantation, pancreatic cells that were transfected with pEBVHGF and then re-aggregated showed ductal cell marker expression. However, ductal cells disappeared and the cells underwent fibrosis in a diabetes mouse model two to five weeks after transplantation; these mice also did not show controlled blood glucose levels. Furthermore, pancreatic cells transplanted into nude mice with normal blood glucose showed poor graft survival regardless of the type of transfected plasmid (pCEP4, pHGF, or pEBVHGF).

Conclusion

For clinical application of transfected neonatal porcine pancreatic cells, further studies are required to develop methods of overcoming the damage for the cells caused by repeated transfection and to re-aggregate them into islet-like structures.

Citations

Citations to this article as recorded by  
  • Successful xenotransplantation with re‐aggregated and encapsulated neonatal pig liver cells for treatment of mice with acute liver failure
    Dong‐Sik Ham, Min‐Sang Song, Heon‐Seok Park, Marie Rhee, Hae Kyung Yang, Seung‐Hwan Lee, Ji‐Won Kim, Eun‐Sun Jung, Kun‐Ho Yoon
    Xenotransplantation.2015; 22(4): 249.     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
  • Prevalence, Awareness, and Control of Hypertension among Diabetic Koreans
    Hyun Hee Chung, Kyu Chang Won
    Diabetes & Metabolism Journal.2011; 35(4): 337.     CrossRef

Diabetes Metab J : Diabetes & Metabolism Journal