Rhein Protects Pancreatic β-Cells From Dynamin-Related Protein-1–Mediated Mitochondrial Fission and Cell Apoptosis Under Hyperglycemia

Diabetes, Nov 2013

Rhein, an anthraquinone compound isolated from rhubarb, has been shown to improve glucose metabolism disorders in diabetic mice. The mechanism underlying the protective effect of rhein, however, remains unknown. Here, we demonstrate that rhein can protect the pancreatic β-cells against hyperglycemia-induced cell apoptosis through stabilizing mitochondrial morphology. Oral administration of rhein for 8 or 16 weeks in db/db mice significantly reduced fasting blood glucose (FBG) level and improved glucose tolerance. Cell apoptosis assay using both pancreatic sections and cultured pancreatic β-cells indicated that rhein strongly inhibited β-cell apoptosis. Morphological study showed that rhein was mainly localized at β-cell mitochondria and rhein could preserve mitochondrial ultrastructure by abolishing hyperglycemia-induced mitochondrial fission protein dynamin-related protein 1 (Drp1) expression. Western blot and functional analysis confirmed that rhein protected the pancreatic β-cells against hyperglycemia-induced apoptosis via suppressing mitochondrial Drp1 level. Finally, mechanistic study further suggested that decreased Drp1 level by rhein might be due to its effect on reducing cellular reactive oxygen species. Taken together, our study demonstrates for the first time that rhein can serve as a novel therapeutic agent for hyperglycemia treatment and rhein protects pancreatic β-cells from apoptosis by blocking the hyperglycemia-induced Drp1 expression.

A PDF file should load here. If you do not see its contents the file may be temporarily unavailable at the journal website or you do not have a PDF plug-in installed and enabled in your browser.

Alternatively, you can download the file locally and open with any standalone PDF reader:

https://diabetes.diabetesjournals.org/content/62/11/3927.full.pdf

Rhein Protects Pancreatic β-Cells From Dynamin-Related Protein-1–Mediated Mitochondrial Fission and Cell Apoptosis Under Hyperglycemia

Jing Liu Zhaohong Chen Yujing Zhang Mingchao Zhang Xiaodong Zhu Yun Fan Shaolin Shi Ke Zen kzen@nju Zhihong Liu Rhein, an anthraquinone compound isolated from rhubarb, has been shown to improve glucose metabolism disorders in diabetic mice. The mechanism underlying the protective effect of rhein, however, remains unknown. Here, we demonstrate that rhein can protect the pancreatic b-cells against hyperglycemia-induced cell apoptosis through stabilizing mitochondrial morphology. Oral administration of rhein for 8 or 16 weeks in db/db mice significantly reduced fasting blood glucose (FBG) level and improved glucose tolerance. Cell apoptosis assay using both pancreatic sections and cultured pancreatic b-cells indicated that rhein strongly inhibited b-cell apoptosis. Morphological study showed that rhein was mainly localized at b-cell mitochondria and rhein could preserve mitochondrial ultrastructure by abolishing hyperglycemiainduced mitochondrial fission protein dynamin-related protein 1 (Drp1) expression. Western blot and functional analysis confirmed that rhein protected the pancreatic b-cells against hyperglycemiainduced apoptosis via suppressing mitochondrial Drp1 level. Finally, mechanistic study further suggested that decreased Drp1 level by rhein might be due to its effect on reducing cellular reactive oxygen species. Taken together, our study demonstrates for the first time that rhein can serve as a novel therapeutic agent for hyperglycemia treatment and rhein protects pancreatic b-cells from apoptosis by blocking the hyperglycemia-induced Drp1 expression. Diabetes 62:3927-3935, 2013 - Racid) is an anthraquinone compound isolated hein (4,5-dihydroxyanthraquinone-2-carboxylic from rhubarb that has been used for more than 2,000 years in China to treat constipation, gastrointestinal hemorrhage, and ulcers (1). In our previous work, we found that rhein could improve glucose metabolism disorders in diabetic mice, and its effect on reducing blood glucose level was even stronger than rosiglitazone and benazepril (2,3). Moreover, rhein also inhibited apoptosis of islet cells and protected islet function (4). Using mouse nonalcoholic fatty liver disease as an animal model associated with obesity, insulin resistance, and inflammatory disorders, Sheng et al. (5) reported that rhein could ameliorate fatty liver disease in diet-induced obese mice via negative energy balance, hepatic lipogenous regulation, and immunomodulation. Recent antihyperglycemic study by Chatterjee et al. (6) suggests that rhein, as well as other natural inhibitors such as aloins and capparisine, may be a foundation for a better antidiabetic therapy. However, the mechanism underlying these protective effects of rhein remains unclear. Increasing evidence suggests that b-cell failure is the mainstay of the pathogenesis of type 2 diabetes (7). Although the precise mechanisms underlying the b-cell dysfunction in type 2 diabetes are not fully understood, hyperglycemia has been shown as a major factor to cause the b-cell apoptosis. Once hyperglycemia develops, the pancreatic b-cell is exposed to increased metabolic flux and associated cellular stress, leading to impairment of b-cell function and survival, a process called glucotoxicity (8,9). In type 2 diabetes, hyperglycemia is commonly associated with deregulation of lipid metabolism and elevation of free fatty acids, which also contribute to b-cell dysfunction (8,10). Moreover, high levels of glucose can also amplify lipotoxicity (10). The thiazolidinedione peroxisome proliferatoractivated receptor-g activator drugs, rosiglitazone and pioglitazone, have been widely used to suppress insulin resistance in type 2 diabetic patients (11). Although rhein shows a similar or even better effect on reducing mouse blood glucose level than rosiglitazone, the underlying mechanism remains unclear. It has been known that mitochondrial fission and fusion modulators, dynaminrelated protein 1 (Drp1) (12), optic atrophy protein 1 (Opa1) (13), prohibitin (14), and mitofusin (15), collectively control the dynamic balance of mitochondria fission and fusion processes and consequent mitochondria functions. Previous studies have demonstrated that Drp1 plays an important role in promoting hyperglycemia-induced apoptosis of b-cells and neurons (12,16,17). Drp1 expression was increased drastically in islet b-cells under hyperglycemia conditions. Estaquier and Arnoult (18) further demonstrated that inhibiting Drp1-mediated mitochondrial fission could selectively prevent the release of cytochrome c, a mediator of apoptosis, from mitochondria. In contrast to the mitochondria fission modulators, which are upregulated or activated by stress factors such as high concentration of glucose (HG), mitochondria fusion modulators are generally reduced when cells are challenged with proapoptotic insults. Recent studies by Kushnareva et al. (19) and Leboucher et al. (15) showed that stress-induced loss of Opa1 and mitofusin can faci (...truncated)


This is a preview of a remote PDF: https://diabetes.diabetesjournals.org/content/62/11/3927.full.pdf

Jing Liu, Zhaohong Chen, Yujing Zhang, Mingchao Zhang, Xiaodong Zhu, Yun Fan, Shaolin Shi, Ke Zen, Zhihong Liu. Rhein Protects Pancreatic β-Cells From Dynamin-Related Protein-1–Mediated Mitochondrial Fission and Cell Apoptosis Under Hyperglycemia, Diabetes, 2013, pp. 3927-3935, 62/11, DOI: 10.2337/db13-0251