1
|
Sahin I, Bakiner O, Demir T, Sari R, Atmaca A. Current Position of Gliclazide and Sulfonylureas in the Contemporary Treatment Paradigm for Type 2 Diabetes: A Scoping Review. Diabetes Ther 2024; 15:1687-1716. [PMID: 38935188 PMCID: PMC11263312 DOI: 10.1007/s13300-024-01612-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
The increasing burden of type 2 diabetes (T2D), in relation to alarming rise in the prevalence; challenges in the diagnosis, prevention, and treatment; as well as the substantial impact of disease on longevity and quality of life, is a major concern in healthcare worldwide. Sulfonylureas (SUs) have been a cornerstone of T2D pharmacotherapy for over 60 years as oral antidiabetic drugs (OADs), while the newer generation SUs, such as gliclazide modified release (MR), are known to be associated with low risk of hypoglycemia in addition to the cardiovascular neutrality. This scoping review aimed to specifically address the current position of gliclazide MR among other SUs in the contemporary treatment paradigm for T2D and to provide a practical guidance document to assist clinicians in using gliclazide MR in real-life clinical practice. The main topics addressed in this paper include the role of early and sustained glycemic control and use of SUs in T2D management, the properties of gliclazide MR in relation to its effectiveness and safety, the use of gliclazide therapy in special populations, and the place of SUs as a class and gliclazide MR specifically in the current T2D treatment algorithm.
Collapse
Affiliation(s)
- Ibrahim Sahin
- Department of Endocrinology and Metabolism, Inonu University Faculty of Medicine, Malatya, Turkey.
| | - Okan Bakiner
- Department of Endocrinology and Metabolism, Baskent University Faculty of Medicine Adana Dr. Turgut Noyan Application and Research Center, Adana, Turkey
| | - Tevfik Demir
- Department of Endocrinology and Metabolism, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Ramazan Sari
- Department of Endocrinology and Metabolism, Akdeniz University Faculty of Medicine, Antalya, Turkey
| | - Aysegul Atmaca
- Department of Endocrinology and Metabolism, Ondokuz Mayis University Faculty of Medicine, Samsun, Turkey
| |
Collapse
|
2
|
Li W, Qiao J, Lin K, Sun P, Wang Y, Peng Q, Ye X, Liu W, Sun B. Ethyl-acetate fraction from a cinnamon-cortex extract protects pancreatic β-cells from oxidative stress damage. Front Pharmacol 2023; 14:1111860. [PMID: 36950010 PMCID: PMC10025376 DOI: 10.3389/fphar.2023.1111860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/24/2023] [Indexed: 03/08/2023] Open
Abstract
Background: The pathogenesis of diabetes mellitus is mediated mainly by oxidative stress produced by damaged pancreatic β-cells. We identified that an ethyl-acetate fraction (EA) from a cinnamon-cortex extract (CCE) is rich in flavonoid, and showed no toxicity to β cells. Objective: In this study, we evaluated the pharmacologic activities of EA on pancreatic β cells using a model of oxidative stress induced by H2O2 or alloxan. Results: The results showed that EA could significantly reduce reactive oxygen (ROS) accumulation to improve the survival of cells. Western blot showed that EA treatment upregulated expression of nuclear factor erythroid 2 related factor 2, heme oxygenase-1, and gamma glutamylcysteine synthetase. The same model study found that EA also can protect β cells against the apoptosis induced by oxidative stress. Furthermore, EA can enhance insulin secretion in rat and mouse β cell lines treated or not with alloxan or H2O2. The expression of the insulin transcription factor PDX-1 increased in an EA concentration-dependent manner. At last, the major functional compounds of EA analysis showed that three compounds, cinnamyl alcohol, coumarin, and cinnamic acid, had similar effects as EA. Conclusions: In sum, our data suggested that EA fraction from CCE can protect β cells from oxidative stress, and increase insulin secretion to improve the function of β cells. This function might be due to these three compounds found in EA. Our findings provide a theoretical basis and functional molecules for the use of CCE against diabetes mellitus.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Wei Liu
- *Correspondence: Binlian Sun, ; Wei Liu,
| | | |
Collapse
|
3
|
Mustapha S, Azemi AK, Wan Ahmad WAN, Rasool AHG, Mustafa MR, Mokhtar SS. Inhibition of Endoplasmic Reticulum Stress Improves Acetylcholine-Mediated Relaxation in the Aorta of Type-2 Diabetic Rats. Molecules 2022; 27:5107. [PMID: 36014347 PMCID: PMC9413505 DOI: 10.3390/molecules27165107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/05/2022] [Accepted: 08/08/2022] [Indexed: 12/02/2022] Open
Abstract
Endoplasmic reticulum (ER) stress contributes to insulin resistance and macro- and microvascular complications associated with diabetes. This study aimed to evaluate the effect of ER stress inhibition on endothelial function in the aorta of type-2 diabetic rats. Type-2 diabetes was developed in male Sprague-Dawley rats using a high-fat diet and low-dose streptozotocin. Rat aortic tissues were harvested to study endothelial-dependent relaxation. The mechanisms for acetylcholine-mediated relaxation were investigated using pharmacological blockers, Western blotting, oxidative stress, and inflammatory markers. Acetylcholine-mediated relaxation was diminished in the aorta of diabetic rats compared to control rats; supplementation with TUDCA improved relaxation. In the aortas of control and diabetic rats receiving TUDCA, the relaxation was mediated via eNOS/PI3K/Akt, NAD(P)H, and the KATP channel. In diabetic rats, acetylcholine-mediated relaxation involved eNOS/PI3K/Akt and NAD(P)H, but not the KATP channel. The expression of ER stress markers was upregulated in the aorta of diabetic rats and reduced with TUDCA supplementation. The expression of eNOS and Akt were lower in diabetic rats but were upregulated after supplementation with TUDCA. The levels of MDA, IL-6, and SOD activity were higher in the aorta of the diabetic rats compared to control rats. This study demonstrated that endothelial function was impaired in diabetes, however, supplementation with TUDCA improved the function via eNOS/Akt/PI3K, NAD(P)H, and the KATP channel. The improvement of endothelial function was associated with increased expressions of eNOS and Akt. Thus, ER stress plays a crucial role in the impairment of endothelial-dependent relaxation. Mitigating ER stress could be a potential strategy for improving endothelial dysfunction in type-2 diabetes.
Collapse
Affiliation(s)
- Sagir Mustapha
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Kelantan, Malaysia
- Department of Pharmacology and Therapeutics, Ahmadu Bello University, Zaria 810107, Kaduna, Nigeria
| | - Ahmad Khusairi Azemi
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Terengganu 21030, Terengganu, Malaysia
| | - Wan Amir Nizam Wan Ahmad
- Biomedicine Programme, School of Health Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Kelantan, Malaysia
| | - Aida Hanum Ghulam Rasool
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Kelantan, Malaysia
| | - Mohd Rais Mustafa
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Selangor, Malaysia
| | - Siti Safiah Mokhtar
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Kelantan, Malaysia
| |
Collapse
|
4
|
Szymczak-Pajor I, Fleszar K, Kasznicki J, Gralewska P, Śliwińska A. A potential role of calpains in sulfonylureas (SUs) -mediated death of human pancreatic cancer cells (1.2B4). Toxicol In Vitro 2021; 73:105128. [PMID: 33652124 DOI: 10.1016/j.tiv.2021.105128] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/01/2021] [Accepted: 02/25/2021] [Indexed: 12/25/2022]
Abstract
Sulfonylureas (SUs) are suggested to accelerate the pancreatic β-cells mass loss via apoptosis. However, little is known whether calpains mediate this process. The aim of the present study is to evaluate the involvement of calpains in SUs-induced death of human pancreatic cancer (PC) cell line 1.2B4. The cells were exposed to: glibenclamide, glimepiride and gliclazide for 72 h. The expression analysis of caspase-3 (CASP-3), TP53, calpain 1 (CAPN-1), calpain 2 (CAPN-2) and calpain 10 (CAPN-10) was detected using RT-PCR method. Intracellular Ca2+ concentrations, CASP-3 activity and total calpain activity were also evaluated. Our results have shown that glibenclamide and glimepiride decrease 1.2B4 cells viability with accompanied increase in intracellular Ca2+ concentration and increased expression of apoptosis-related CASP-3 and TP53. Gliclazide did not affect 1.2B4 cell viability and Ca2+ concentration, however, it downregulated CASP-3 and upregulated TP53. Interestingly, 50 μM glimepiride increased expression of CAPN-1, CAPN-2 and CAPN-10 whereas 50 μM glibenclamide solely upregulated CAPN-2 expression. We have shown that 10 μM and 50 μM glibenclamide and glimepiride increased the activity of CASP-3, but decreased total calpain activity. Our results suggest that calpains may be involved in glibenclamide- and glimepiride-induced death of PC cells. However, further investigation is required to confirm the engagement of calpains in SUs-mediated death of PC cells, especially studies on protein level of particular isoforms of calpains should be conducted.
Collapse
Affiliation(s)
- Izabela Szymczak-Pajor
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska, 92-213 Lodz, Poland.
| | - Krzysztof Fleszar
- Student Scientific Society of Civilization Diseases, Medical University of Lodz, 251 Pomorska, 92-213 Lodz, Poland.
| | - Jacek Kasznicki
- Department of Internal Diseases, Diabetology and Clinical Pharmacology, Medical University of Lodz, 251 Pomorska, 92-213 Lodz, Poland.
| | - Patrycja Gralewska
- Department of Medical Biophysics, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland.
| | - Agnieszka Śliwińska
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska, 92-213 Lodz, Poland.
| |
Collapse
|
5
|
Jahan H, Choudhary MI. Gliclazide alters macrophages polarization state in diabetic atherosclerosis in vitro via blocking AGE-RAGE/TLR4-reactive oxygen species-activated NF-kβ nexus. Eur J Pharmacol 2021; 894:173874. [PMID: 33460615 DOI: 10.1016/j.ejphar.2021.173874] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/07/2021] [Accepted: 01/11/2021] [Indexed: 12/17/2022]
Abstract
Hyperglycemic milieu in diabetes mellitus stimulates macrophages for exaggerated pro-inflammatory cytokine response, particularly IL-1β, IL-6, and TNF-α. Although hyperglycemia causes macrophages to produce pro-inflammatory cytokines, AGEs (advanced glycation end products) active inflammation, produced as a result of chronic hyperglycemia, inducers cause polarization of macrophages into pro-inflammatory M1 phenotype. AGEs in diabetes accelerate atherosclerotic plaque initiation and progression via promoting macrophages polarization towards pro-inflammatory state. Gliclazide (Glz) is a well known antidiabetic drug with excellent safety profile. Its repurposing in the management of diabetes-associated late complications has tremendous merit. The present study demonstrated that Glz retards diabetic atherosclerotic progression, and cytokines storm in a concentration dependent manner over a concentration range of 1-100 μM than those of AGEs (200 μg/ml)-treated cells through a mechanism that alters macrophage M1 polarization state. Glz exerted these beneficial effects, independent of its antidiabetic effect. Glz pretreatment significantly (P < 0.05) inhibited the AGEs-induced pro-inflammatory mediators (NO•, reactive oxygen species, i-NOS), and production of pro-inflammatory cytokines, including IL-1β, IL-6, and TNF-α. It also significantly (P < 0.05) promoted the production of anti-inflammatory cytokines (IL-10 and TGF-β) in RAW 264.7 mouse macrophages. Glz pretreatment also effectively abated the AGEs-induced RAGE (~2-fold decrease), and CD86 surface marker expressions (P < 0.001 at 100 μM) on macrophages by inhibiting the NF-kβ activation in a concentration dependent manner (1-100 μM) (P < 0.001). In conclusion, our data demonstrates that Glz alleviates the diabetic atherosclerosis progression by ameliorating the AGEs-mediated M1 pro-inflammatory phenotype via blocking AGE-RAGE/TLR4-reactive oxygen species -activated NF-kβ nexus in macrophages.
Collapse
Affiliation(s)
- Humera Jahan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Science, University of Karachi, Karachi, 75270, Pakistan.
| | - M Iqbal Choudhary
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Science, University of Karachi, Karachi, 75270, Pakistan; H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Science, University of Karachi, Karachi, 75270, Pakistan; Department of Chemistry, Faculty of Science and Technology, Universitas Airlangga, Komplek Kampus C, JI. Mulyorejo, Surabaya, 60115, Indonesia.
| |
Collapse
|
6
|
Network Pharmacology-Based Strategy Reveals the Effects of Hedysarum multijugum Maxim.- Radix Salviae Compound on Oxidative Capacity and Cardiomyocyte Apoptosis in Rats with Diabetic Cardiomyopathy. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8260703. [PMID: 33134388 PMCID: PMC7591987 DOI: 10.1155/2020/8260703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 08/04/2020] [Indexed: 11/25/2022]
Abstract
Objective To explore the effects of the Hedysarum multijugum Maxim.-Radix Salviae compound (Huangqi-Danshen Compound (HDC)) on oxidative capacity and cardiomyocyte apoptosis in rats with diabetic cardiomyopathy by a network pharmacology-based strategy. Methods Traditional Chinese Medicine (TCM)@Taiwan, TCM Systems Pharmacology Database and Analysis Platform (TCMSP), TCM Integrated Database (TCMID), and High-Performance Liquid Chromatography (HPLC) technology were used to obtain and screen HDC's active components, and the PharmMapper database was used to predict HDC human target protein targets. The DCM genes were collected from the GeneCards and OMIM databases, and the network was constructed and analyzed by Cytoscape 3.7.1 and the Database for Annotation, Visualization, and Integrated Discovery (DAVID). Finally, HDC was used to intervene in diabetic cardiomyopathy (DCM) model rats, and important biological processes and signaling pathways were verified using techniques such as immunohistochemistry. Results A total of 176 of HDC's active components and 442 potential targets were obtained. The results of network analysis show that HDC can regulate DCM-related biological processes (such as negative regulation of the apoptotic process, response to hypoxia, the steroid hormone-mediated signaling pathway, cellular iron ion homeostasis, and positive regulation of phosphatidylinositol 3-kinase signaling) and signaling pathways (such as the HIF-1 signaling pathway, the estrogen signaling pathway, insulin resistance, the PPAR signaling pathway, the VEGF signaling pathway, and the PI3K-Akt signaling pathway). Animal experiments show that HDC can reduce fasting plasma glucose (FPG), HbA1c, and malondialdehyde (MDA) and increase superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) (P < 0.05). The results of immunohistochemistry showed that HDC can regulate the protein expression of apoptosis-related signaling pathways in DCM rats (P < 0.05). Conclusion It was initially revealed that HDC improves DCM through its antiapoptotic and anti-inflammatory effects. HDC may play a therapeutic role by improving cardiomyocyte apoptosis in DCM rats.
Collapse
|
7
|
Luppi P, Drain N, To R, Stolz D, Wallace C, Watkins S, Drain P. Autocrine C-peptide protects INS1 β cells against palmitic acid-induced oxidative stress in peroxisomes by inducing catalase. Endocrinol Diabetes Metab 2020; 3:e00147. [PMID: 32704568 PMCID: PMC7375117 DOI: 10.1002/edm2.147] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/26/2020] [Accepted: 05/02/2020] [Indexed: 12/12/2022] Open
Abstract
AIMS C-peptide, produced by pancreatic β cells and co-secreted in the bloodstream with insulin, has antioxidant properties in glucose- and hydrogen peroxide (H2O2)-exposed INS1 β cells. Palmitic acid, the most physiologically abundant long-chain free fatty acid in humans, is metabolized in peroxisomes of β cells accumulating H2O2 that can lead to oxidative stress. Here, we tested the hypothesis that C-peptide protects β cells from palmitic acid-induced stress by lowering peroxisomal H2O2. MATERIALS AND METHODS We exposed INS1 β cells to palmitic acid and C-peptide in the setting of increasing glucose concentration and tested for changes in parameters of stress and death. To study the ability of C-peptide to lower peroxisomal H2O2, we engineered an INS1 β cell line stably expressing the peroxisomal-targeted H2O2 sensor HyPer, whose fluorescence increases with cellular H2O2. An INS1 β cell line stably expressing a live-cell fluorescent catalase reporter was used to detect changes in catalase gene expression. RESULTS C-peptide protects INS1 β cells from the combined effect of palmitic acid and glucose by reducing peroxisomal H2O2 to baseline levels and increasing expression of catalase. CONCLUSIONS In conditions of glucolipotoxicity, C-peptide increases catalase expression and reduces peroxisomal oxidative stress and death of INS1 β cells. Maintenance of C-peptide secretion is a pro-survival requisite for β cells in adverse conditions. Loss of C-peptide secretion would render β cells more vulnerable to stress and death leading to secretory dysfunction and diabetes.
Collapse
Affiliation(s)
- Patrizia Luppi
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Nicholas Drain
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Ramsey To
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Donna Stolz
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Callen Wallace
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Simon Watkins
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Peter Drain
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| |
Collapse
|
8
|
Hendriks AM, Schrijnders D, Kleefstra N, de Vries EGE, Bilo HJG, Jalving M, Landman GWD. Sulfonylurea derivatives and cancer, friend or foe? Eur J Pharmacol 2019; 861:172598. [PMID: 31408647 DOI: 10.1016/j.ejphar.2019.172598] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/07/2019] [Accepted: 08/08/2019] [Indexed: 02/07/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is associated with a higher risk of cancer and cancer-related mortality. Increased blood glucose and insulin levels in T2DM patients may be, at least in part, responsible for this effect. Indeed, lowering glucose and/or insulin levels pharmacologically appears to reduce cancer risk and progression, as has been demonstrated for the biguanide metformin in observational studies. Studies investigating the influence of sulfonylurea derivatives (SUs) on cancer risk have provided conflicting results, partly due to comparisons with metformin. Furthermore, little attention has been paid to within-class differences in systemic and off-target effects of the SUs. The aim of this systematic review is to discuss the available preclinical and clinical evidence on how the different SUs influence cancer development and risk. Databases including PubMed, Cochrane, Database of Abstracts on Reviews and Effectiveness, and trial registries were systematically searched for available clinical and preclinical evidence on within-class differences of SUs and cancer risk. The overall preclinical and clinical evidence suggest that the influence of SUs on cancer risk in T2DM patients differs between the various SUs. Potential mechanisms include differing affinities for the sulfonylurea receptors and thus differential systemic insulin exposure and off-target anti-cancer effects mediated for example through potassium transporters and drug export pumps. Preclinical evidence supports potential anti-cancer effects of SUs, which are of interest for further studies and potentially repurposing of SUs. At this time, the evidence on differences in cancer risk between SUs is not strong enough to guide clinical decision making.
Collapse
Affiliation(s)
- Anne M Hendriks
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Dennis Schrijnders
- Langerhans Medical Research Group, Zwolle, the Netherlands; Diabetes Center, Isala Hospital, Zwolle, the Netherlands
| | | | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Henk J G Bilo
- Diabetes Center, Isala Hospital, Zwolle, the Netherlands; Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Mathilde Jalving
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Gijs W D Landman
- Langerhans Medical Research Group, Zwolle, the Netherlands; Department of Internal Medicine, Gelre Hospital, Apeldoorn, the Netherlands
| |
Collapse
|
9
|
Yao H, Feng J, Zheng Q, Wei Y, Yang G, Feng W. Comparison of the Effects of Prophylactic and Therapeutic Administrations on Peripheral Neuropathy in Streptozotocin-Diabetic Rats with Gliclazide or Methylcobalamin. Exp Clin Endocrinol Diabetes 2018; 128:635-643. [PMID: 30453342 DOI: 10.1055/a-0635-0672] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
OBJECTIVE To observe the differences in curative effects between prophylactic and therapeutic administrations of Gliclazide (GLZ) or Methylcobalamin (MCA) on diabetic peripheral neuropathy in rats. METHODS GLZ (25 mg/kg/day) or MCA (175 μg/kg/day) was orally administrated prophylactically to streptozotocin-induced diabetic rats for 8 weeks before diabetic peripheral neuropathy developed or administrated therapeutically after diabetic peripheral neuropathy developed, respectively. The motor nerve conduction velocities (MNCV), aldose reductase (AR) activities, the polyol contents and antioxidative enzyme activities in the sciatic never tissues were determined. The morphology of sciatic never tissues was observed. RESULTS In comparison to vehicle, most of the changes in the sciatic nerves of the diabetic rats (e. g., delayed MNCV, altered/damaged nerve structure, enhanced AR activity, increased polyol contents, altered Cu, Zn-superoxide dismutase, glutathione-peroxidase activities, and elevated malondialdehyde level) were significantly ameliorated by prophylactic administration with either GLZ or MCA. In contrast, only few of above-mentioned parameters were alleviated in DPN rats by therapeutic administration with GLZ or MCA as compared to vehicle. The curative effects of GLZ or MCA prophylactic administration on MNCV, AR activity, polyol contents and antioxidative enzyme activities were markedly stronger than therapeutic administration. CONCLUSION Prophylactic administration of GLZ or MCA was superior to the therapeutic administration in alleviation of diabetic neuropathy in STZ-rats, suggesting that pharmacotherapy should be initiated at a much earlier stage before diabetic neuropathy developed, but not at a later stage after never damage reached.
Collapse
Affiliation(s)
- Hongping Yao
- First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, China
| | - Juanyi Feng
- Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, China
| | - Qiaowei Zheng
- First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, China
| | - Youxia Wei
- First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, China
| | - Guangde Yang
- Department of Pharmacology, Medical College of Xi'an Jiaotong University, Xi'an, China
| | - Weiyi Feng
- First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
10
|
Long B, Gan TY, Zhang RC, Zhang YH. miR-23a Regulates Cardiomyocyte Apoptosis by Targeting Manganese Superoxide Dismutase. Mol Cells 2017; 40:542-549. [PMID: 28756653 PMCID: PMC5582300 DOI: 10.14348/molcells.2017.0012] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 06/22/2017] [Accepted: 06/25/2017] [Indexed: 02/06/2023] Open
Abstract
Cardiomyocyte apoptosis is initiated by various cellular insults and accumulated cardiomyocyte apoptosis leads to the pathogenesis of heart failure. Excessive reactive oxygen species (ROS) provoke apoptotic cascades. Manganese superoxide dismutase (MnSOD) is an important antioxidant enzyme that converts cellular ROS into harmless products. In this study, we demonstrate that MnSOD is down-regulated upon hydrogen peroxide treatment or ischemia/reperfusion (I/R) injury. Enhanced expression of MnSOD attenuates cardiomyocyte apoptosis and myocardial infarction induced by I/R injury. Further, we show that miR-23a directly regulates the expression of MnSOD. miR-23a regulates cardiomyocyte apoptosis by suppressing the expression of MnSOD. Our study reveals a novel model regulating cardiomyocyte apoptosis which is composed of miR-23a and MnSOD. Our study provides a new method to tackling apoptosis related cardiac diseases.
Collapse
Affiliation(s)
- Bo Long
- Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730,
China
| | - Tian-Yi Gan
- State Key Laboratory of Cardiovascular Disease, Heart Failure center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100037,
China
| | - Rong-Cheng Zhang
- State Key Laboratory of Cardiovascular Disease, Heart Failure center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100037,
China
| | - Yu-Hui Zhang
- State Key Laboratory of Cardiovascular Disease, Heart Failure center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100037,
China
| |
Collapse
|
11
|
Abstract
Gliclazide is a second-generation oral hypoglycemic drug used for the treatment of noninsulin-dependent diabetes mellitus. It belongs to the sulfonylurea class that stimulates insulin secretion from pancreatic β-cells by inhibiting ATP-dependent potassium channels. Gliclazide also possesses unique antioxidant properties and other beneficial hemobiological effects. This profile represents a comprehensive description of the physical properties, chemical synthesis, spectroscopic characterization (FTIR, 1H NMR, 13C NMR, UV, and single-crystal X-ray), methods of analysis, pharmacological actions, and pharmacokinetic and pharmacodynamic properties of the title drug.
Collapse
|
12
|
Gupta P, Bala M, Gupta S, Dua A, Dabur R, Injeti E, Mittal A. Efficacy and risk profile of anti-diabetic therapies: Conventional vs traditional drugs—A mechanistic revisit to understand their mode of action. Pharmacol Res 2016; 113:636-674. [DOI: 10.1016/j.phrs.2016.09.029] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 09/23/2016] [Accepted: 09/23/2016] [Indexed: 12/17/2022]
|
13
|
Abstract
OBJECTIVES Mechanisms of toxicity and cell damage were investigated in novel clonal human pancreatic beta cell line, 1.1B4, after exposure to streptozotocin, alloxan, ninhydrin, and hydrogen peroxide. METHODS Viability, DNA damage, insulin secretion/content, [Ca]i, and glucokinase/hexokinase, mRNA expression were measured by MTT assay, comet assay, radioimmunoassay, fluorometric imaging plate reader, enzyme-coupled photometry, and real-time polymerase chain reaction, respectively. RESULTS Chemicals significantly reduced 1.1B4 cell viability in a time/concentration-dependent manner. Chronic 18-hour exposure decreased cellular insulin, glucokinase, and hexokinase activities. Chemicals decreased transcription of INS, GCK, PCSK1, PCSK2, and GJA1 (involved in secretory function). Insulin release and [Ca]i responses to nutrients and membrane-depolarizing agents were impaired. Streptozotocin and alloxan up-regulated transcription of genes, SOD1 and SOD2 (antioxidant enzymes). Ninhydrin and hydrogen peroxide up-regulated SOD2 transcription, whereas alloxan and hydrogen peroxide increased CAT transcription. Chemicals induced DNA damage, apoptosis, and increased caspase 3/7 activity. Streptozotocin and alloxan decreased transcription of BCL2 while increasing transcription of BAX. Chemicals did not affect transcription of HSPA4 and HSPA5 and nitrite production. CONCLUSIONS 1.1B4 cells represent a useful model of human beta cells. Chemicals impaired 1.1B4 cell secretory function and activated antioxidant defense and apoptotic pathways without activating endoplasmic reticulum stress response/nitrosative stress.
Collapse
|
14
|
Sim YC, Lee JS, Lee S, Son YK, Park JE, Song JE, Ha SJ, Hong EK. Effects of polysaccharides isolated from Inonotus obliquus against hydrogen peroxide-induced oxidative damage in RINm5F pancreatic β-cells. Mol Med Rep 2016; 14:4263-4270. [DOI: 10.3892/mmr.2016.5763] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 08/18/2016] [Indexed: 11/06/2022] Open
|
15
|
Yao H, Feng J, Zheng Q, Wei Y, Wang S, Feng W. The effects of gliclazide, methylcobalamin, and gliclazide+methylcobalamin combination therapy on diabetic peripheral neuropathy in rats. Life Sci 2016; 161:60-8. [DOI: 10.1016/j.lfs.2016.07.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 07/18/2016] [Accepted: 07/30/2016] [Indexed: 11/25/2022]
|
16
|
Chan SP, Colagiuri S. Systematic review and meta-analysis of the efficacy and hypoglycemic safety of gliclazide versus other insulinotropic agents. Diabetes Res Clin Pract 2015; 110:75-81. [PMID: 26361859 DOI: 10.1016/j.diabres.2015.07.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 06/17/2015] [Accepted: 07/02/2015] [Indexed: 12/12/2022]
Abstract
AIMS Sulfonylureas are well positioned in treating type 2 diabetes, after lifestyle modification and metformin. The sulfonylurea gliclazide was given preference over glibenclamide in older people with type 2 diabetes in the World Health Organization model list of essential medicines. Consequently, a systematic review and meta-analysis of randomized controlled trials of the efficacy and safety of gliclazide versus other oral insulinotropic agents (sulfonylureas, dipeptidyl peptidase-4 inhibitors, and glinides) was performed. METHODS Two reviewers searched MEDLINE for studies of ≥12 weeks duration in adults with type 2 diabetes. The key search word was "gliclazide", filtered with "randomized controlled trial", "human" and "19+ years". Differences were explored in mean change in glycated hemoglobin (HbA(1c)) from baseline (primary outcome) and risk of hypoglycemia (secondary outcome) between gliclazide and other oral insulinotropic agents; and other sulfonylureas. RESULTS Nine out of 181 references reported primary outcomes, of which 7 reported secondary outcomes. Gliclazide lowered HbA1c more than other oral insulinotropic agents, with a weighted mean difference of -0.11% (95%, CI -0.19 to -0.03%, P=0.008, I(2)=60%), though not more than other sulfonylureas (-0.12%; 95%, CI -0.25 to 0.01%, P=0.07, I(2)=77%). Risk of hypoglycemia with gliclazide was not different to other insulinotropic agents (RR 0.85; 95%, CI 0.66 to 1.09, P=0.20, I(2)=61%) but significantly lower than other sulfonylureas (RR 0.47; 95%, CI 0.27 to 0.79, P=0.004, I(2)=0%). CONCLUSION Compared with other oral insulinotropic agents, gliclazide significantly reduced HbA1c with no difference regarding hypoglycemia risk. Compared with other sulfonylureas, HbA1c reduction with gliclazide was not significantly different, but hypoglycemia risk was significantly lower.
Collapse
Affiliation(s)
- Siew Pheng Chan
- Department of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| | | |
Collapse
|
17
|
Weaver JD, Stabler CL. Antioxidant cerium oxide nanoparticle hydrogels for cellular encapsulation. Acta Biomater 2015; 16:136-44. [PMID: 25620795 DOI: 10.1016/j.actbio.2015.01.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 12/10/2014] [Accepted: 01/13/2015] [Indexed: 10/24/2022]
Abstract
Oxidative stress and the resulting radical by-products cause significant toxicity and graft loss in cellular transplantation. Here, the engineering of an auto-catalytic, antioxidant, self-renewing cerium oxide nanoparticle (CONP)-composite hydrogel is reported. This enzyme-mimetic material ubiquitously scavenges ambient free radicals, with the potential to provide indefinite antioxidant protection. The potential of this system to enhance the protection of encapsulated beta cells was evaluated. Co-incubation of CONPs free in solution with beta cells demonstrated potent cytoprotection from superoxide exposure; however, phagocytosis of the CONPs by the beta cells resulted in cytotoxicity at concentrations as low as 1mM. When CONPs were embedded within alginate hydrogels, the composite hydrogel provided cytoprotection to encapsulated beta cells from free radical attack without cytotoxicity, even up to 10mM. This nanocomposite hydrogel has wide applicability in cellular transplantation, with the unique advantage of localization of these potent antioxidant CONPs and their capacity for sustained, long-term scavenging.
Collapse
|
18
|
Mogensen UM, Andersson C, Fosbøl EL, Schramm TK, Vaag A, Scheller NM, Torp-Pedersen C, Gislason G, Køber L. Metformin in combination with various insulin secretagogues in type 2 diabetes and associated risk of cardiovascular morbidity and mortality--a retrospective nationwide study. Diabetes Res Clin Pract 2015; 107:104-12. [PMID: 25458330 DOI: 10.1016/j.diabres.2014.09.047] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 08/21/2014] [Accepted: 09/15/2014] [Indexed: 01/13/2023]
Abstract
AIMS Metformin is the first-line treatment for most patients with type 2 diabetes but many patients need additional treatment with insulin secretagogues (IS) to achieve glycemic control. We aimed to compare mortality and cardiovascular risk among users of metformin in combination with pharmacologically different ISs. METHODS Using nationwide administrative Danish registries, we followed all individuals without prior stroke or myocardial infarction who initiated metformin and an IS from 1997 through 2009. Rate ratios (RR) of all-cause mortality, cardiovascular death, and a composite of myocardial infarction, stroke, or cardiovascular death were compared between user groups using time-dependent multivariable Poisson regression models. The most common combination, glimepiride+metformin, was used as reference. RESULTS A total of 56,827 patients were included, 56% male, the mean age was 61 ± 12.5 years, and median duration of prior monotherapy was 2.2 (inter quartile range 0.5-4.5) years. Crude incidence rates of mortality for combinations of ISs with metformin were; 15.4 (repaglinide), 28.1 (glipizide), 23.7 (glibenclamide), 21.1 (gliclazide), 20.7 (glimepiride), 27.7 (tolbutamide) deaths per 1000 person years. In adjusted analysis, the associated mortality risk was similar for users of gliclazide+metformin (RR=1.01 [0.88-1.15]), repaglinide+metformin (RR=0.81 [0.62-1.05]), glibenclamide+metformin (RR=0.98 [0.87-1.10]), and tolbutamide+metformin (RR=1.04 [0.85-1.28]). Users of glipizide+metformin was associated with increased all-cause mortality (RR=1.16 [1.02-1.32], p=0.02), cardiovascular death (RR=1.21 [1.01-1.46], p=0.04), and the combined endpoint (RR=1.20 [1.06-1.36, p=0.005). CONCLUSION Most ISs in combination with metformin were associated with similar mortality and cardiovascular risk. Whether glipizide is associated with increased risk compared with other ISs when used in combinations with metformin warrants further study.
Collapse
Affiliation(s)
| | | | - Emil Loldrup Fosbøl
- Department of Cardiology, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Tina Ken Schramm
- Department of Cardiology, University Hospital Frederiksberg, Copenhagen, Denmark
| | - Allan Vaag
- Department of Endocrinology, University Hospital Rigshospitalet, Copenhagen, Denmark
| | | | | | - Gunnar Gislason
- Department of Cardiology, University Hospital Gentofte, Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; National Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | - Lars Køber
- Department of Cardiology, University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
19
|
Yosten GLC, Maric-Bilkan C, Luppi P, Wahren J. Physiological effects and therapeutic potential of proinsulin C-peptide. Am J Physiol Endocrinol Metab 2014; 307:E955-68. [PMID: 25249503 PMCID: PMC4254984 DOI: 10.1152/ajpendo.00130.2014] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Connecting Peptide, or C-peptide, is a product of the insulin prohormone, and is released with and in amounts equimolar to those of insulin. While it was once thought that C-peptide was biologically inert and had little biological significance beyond its role in the proper folding of insulin, it is now known that C-peptide binds specifically to the cell membranes of a variety of tissues and initiates specific intracellular signaling cascades that are pertussis toxin sensitive. Although it is now clear that C-peptide is a biologically active molecule, controversy still remains as to the physiological significance of the peptide. Interestingly, C-peptide appears to reverse the deleterious effects of high glucose in some tissues, including the kidney, the peripheral nerves, and the vasculature. C-peptide is thus a potential therapeutic agent for the treatment of diabetes-associated long-term complications. This review addresses the possible physiologically relevant roles of C-peptide in both normal and disease states and discusses the effects of the peptide on sensory nerve, renal, and vascular function. Furthermore, we highlight the intracellular effects of the peptide and present novel strategies for the determination of the C-peptide receptor(s). Finally, a hypothesis is offered concerning the relationship between C-peptide and the development of microvascular complications of diabetes.
Collapse
Affiliation(s)
- Gina L C Yosten
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, St. Louis, Missouri;
| | - Christine Maric-Bilkan
- Division of Cardiovascular Sciences, Vascular Biology and Hypertension Branch, National Heart, Lung and Blood Institute, Bethesda, Maryland; Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Patrizia Luppi
- Department of Cell Biology, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania
| | - John Wahren
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; and Cebix Inc., Karolinska Institutet Science Park, Solna, Sweden
| |
Collapse
|
20
|
Lasram MM, Dhouib IB, Annabi A, El Fazaa S, Gharbi N. A review on the molecular mechanisms involved in insulin resistance induced by organophosphorus pesticides. Toxicology 2014; 322:1-13. [DOI: 10.1016/j.tox.2014.04.009] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 04/23/2014] [Accepted: 04/24/2014] [Indexed: 02/06/2023]
|
21
|
Nakajima S, Kitamura M. Bidirectional regulation of NF-κB by reactive oxygen species: a role of unfolded protein response. Free Radic Biol Med 2013; 65:162-174. [PMID: 23792277 DOI: 10.1016/j.freeradbiomed.2013.06.020] [Citation(s) in RCA: 225] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2013] [Revised: 06/06/2013] [Accepted: 06/07/2013] [Indexed: 12/15/2022]
Abstract
Nuclear factor-κB (NF-κB) is a transcription factor that plays a crucial role in coordinating innate and adaptive immunity, inflammation, and apoptotic cell death. NF-κB is activated by various inflammatory stimuli including peptide factors and infectious microbes. It is also known as a redox-sensitive transcription factor activated by reactive oxygen species (ROS). Over the past decades, various investigators focused on the role of ROS in the activation of NF-κB by cytokines and lipopolysaccharides. However, recent studies also suggested that ROS have the potential to repress NF-κB activity. Currently, it is not well addressed how ROS regulate activity of NF-κB in a bidirectional fashion. In this paper, we summarize evidence for positive and negative regulation of NF-κB by ROS, possible redox-sensitive targets for NF-κB signaling, and mechanisms underlying biphasic and bidirectional influences of ROS on NF-κB, especially focusing on a role of ROS-mediated induction of endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Shotaro Nakajima
- Department of Molecular Signaling, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Shimokato 1110, Chuo, Yamanashi 409-3898, Japan
| | - Masanori Kitamura
- Department of Molecular Signaling, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Shimokato 1110, Chuo, Yamanashi 409-3898, Japan.
| |
Collapse
|
22
|
Pasello G, Urso L, Conte P, Favaretto A. Effects of sulfonylureas on tumor growth: a review of the literature. Oncologist 2013; 18:1118-25. [PMID: 24043597 DOI: 10.1634/theoncologist.2013-0177] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Type 2 diabetes mellitus patients are at higher cancer risk, probably because of hyperinsulinemia and insulin growth factor 1 pathway activation. The effects of antidiabetic drugs on cancer risk have been described and discussed in several studies suggesting opposite effects of the biguanide metformin and sulfonylureas on cancer incidence and mortality. The anticancer mechanisms of metformin have been clarified, and some clinical studies, particularly in breast cancer patients, have been published or are currently ongoing; however, data about the effects of sulfonylureas on cancer growth are less consistent. The aims of this work are to review preclinical evidence of second-generation sulfonylureas effects on tumor growth, to clarify the potential mechanisms of action, and to identify possible metabolic targets for patient selection. Most evidence is on the adenosine triphosphate-sensitive potassium channels inhibitor glibenclamide, which interacts with reactive oxygen species production thus inducing cancer cell death. Among diarylsulfonylureas, next-generation DW2282 derivatives are particularly promising because of the proapoptotic activity in multidrug-resistant cells.
Collapse
Affiliation(s)
- Giulia Pasello
- Second Medical Oncology Unit, Istituto Oncologico Veneto, Padua, Italy
| | | | | | | |
Collapse
|
23
|
Bensellam M, Laybutt DR, Jonas JC. The molecular mechanisms of pancreatic β-cell glucotoxicity: recent findings and future research directions. Mol Cell Endocrinol 2012; 364:1-27. [PMID: 22885162 DOI: 10.1016/j.mce.2012.08.003] [Citation(s) in RCA: 208] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 07/11/2012] [Accepted: 08/01/2012] [Indexed: 02/06/2023]
Abstract
It is well established that regular physiological stimulation by glucose plays a crucial role in the maintenance of the β-cell differentiated phenotype. In contrast, prolonged or repeated exposure to elevated glucose concentrations both in vitro and in vivo exerts deleterious or toxic effects on the β-cell phenotype, a concept termed as glucotoxicity. Evidence indicates that the latter may greatly contribute to the pathogenesis of type 2 diabetes. Through the activation of several mechanisms and signaling pathways, high glucose levels exert deleterious effects on β-cell function and survival and thereby, lead to the worsening of the disease over time. While the role of high glucose-induced β-cell overstimulation, oxidative stress, excessive Unfolded Protein Response (UPR) activation, and loss of differentiation in the alteration of the β-cell phenotype is well ascertained, at least in vitro and in animal models of type 2 diabetes, the role of other mechanisms such as inflammation, O-GlcNacylation, PKC activation, and amyloidogenesis requires further confirmation. On the other hand, protein glycation is an emerging mechanism that may play an important role in the glucotoxic deterioration of the β-cell phenotype. Finally, our recent evidence suggests that hypoxia may also be a new mechanism of β-cell glucotoxicity. Deciphering these molecular mechanisms of β-cell glucotoxicity is a mandatory first step toward the development of therapeutic strategies to protect β-cells and improve the functional β-cell mass in type 2 diabetes.
Collapse
Affiliation(s)
- Mohammed Bensellam
- Université catholique de Louvain, Institut de recherche expérimentale et clinique, Pôle d'endocrinologie, diabète et nutrition, Brussels, Belgium
| | | | | |
Collapse
|
24
|
Abstract
The importance of K(ATP) channels in stimulus-secretion coupling of β-cells is well established, although they are not indispensable for the maintenance of glycaemic control. This review article depicts a new role for K(ATP) channels by showing that genetic or pharmacological ablation of these channels protects β-cells against oxidative stress. Increased production of oxidants is a crucial factor in the pathogenesis of type 2 diabetes mellitus (T2DM). T2DM develops when β-cells can no longer compensate for the high demand of insulin resulting from excess fuel intake. Instead β-cells start to secrete less insulin and β-cell mass is diminished by apoptosis. Both, reduction of insulin secretion and β-cell mass induced by oxidative stress, are prevented by deletion or inhibition of K(ATP) channels. These findings may open up new insights into the early treatment of T2DM.
Collapse
Affiliation(s)
- G Drews
- Department of Pharmacology, Institute of Pharmacy, University of Tübingen, Tübingen, Germany.
| | | |
Collapse
|
25
|
Felipe ET, Maestri JS, Kanunfre CC, Curi R, Newsholme P, Carpinelli AR, Oliveira-Emilio HR. Cytotoxicity and cytoprotective effects of citrus flavonoids on insulin-secreting cells BRIN-BD11: beneficial synergic effects. Nat Prod Res 2012; 27:925-8. [PMID: 22443082 DOI: 10.1080/14786419.2012.671315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Flavonoids, in general, have potent antioxidant activity and they can be used in treating chronic diseases involving oxidative stress, such as diabetes mellitus. The purpose of this study was to evaluate the cytotoxicity and cytoprotective effects of citrus flavonoids on the functionality of BRIN-BD11 cells. The assessment of cytotoxic and cytoprotective flavonoid tested was performed using the MTT reduction assay. The flavonoids did not show cytotoxic effects in any of the tested concentrations (5-20 µM) and also negative insulinotropic effects were not observed. To cytoprotective assay, the IC50 of H2O2 in treatment of 2 h (acute oxidative stress) was measured (350 µM). Moreover, under acute oxidative stress, the isolated flavonoids (10 µM) had no cytoprotective effects. Besides an antioxidant role of the flavonoids was only observed when using in association. Thus future experiments are needed, varying the experimental condition, to better evaluate the possible mechanisms of action of these flavonoids.
Collapse
Affiliation(s)
- E T Felipe
- Evolutive Biology Post-Graduate Course, Ponta Grossa State University, Ponta Grossa, Parana, Brazil
| | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Type 2 diabetes mellitus (T2DM) is a progressive disease characterized by worsening hyperglycaemia. Lowering haemoglobin A1c to below or around 7% has been shown to reduce microvascular and neuropathic complications of diabetes. The ongoing uncertainty regarding whether intensive glycaemic control can reduce the increased risk of cardiovascular disease (CVD) in people with T2DM stirred the launch of the recent long-term megatrials. These trials compared the effects of intensive vs. standard control on vascular complications in relatively high CV risk participants with T2DM. While in Veterans Affairs Diabetes Trial, and Action to Control Cardiovascular Risk in Diabetes, the effect of glucose optimization resulted either in no protection or in an excessive CVD death, the Action in Diabetes and Vascular Disease: Preterax and Diamicron Modified Release Controlled Evaluation trial showed that intensive glycaemic control reduced the risk of combined major macrovascular and microvascular events. In this trial, the glucose control strategy was based on gliclazide MR at randomization in all patients and then further sequential addition of other glucose-lowering drugs. Several studies showed that gliclazide has antioxidant properties, reduces markers of endothelial inflammation, and prevents glucose-induced apoptosis of endothelial cells. These positive antioxidant effects are not confined to the vascular wall but they are effective also in the β cells. These properties are important because (i) in patients with atherosclerotic process, microvascular abnormalities may hasten disease progression and (ii) slowing the microvascular complications may have a potentially remarkable effect on the natural history of macrovascular disease.
Collapse
Affiliation(s)
- A Avogaro
- Department of Clinical and Experimental Medicine, University of Padova, Venetian Institute of Molecular Medicine, Padova, Italy.
| |
Collapse
|
27
|
Seino S, Takahashi H, Takahashi T, Shibasaki T. Treating diabetes today: a matter of selectivity of sulphonylureas. Diabetes Obes Metab 2012; 14 Suppl 1:9-13. [PMID: 22118705 DOI: 10.1111/j.1463-1326.2011.01507.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
It is well known that sulphonylureas (SUs), commonly used in the treatment of type 2 diabetes mellitus, stimulate insulin secretion by closing ATP-sensitive K(+) (K(ATP) ) channels in pancreatic β-cells by binding to the SU receptor SUR1. SUs are now known also to activate cAMP sensor Epac2 (cAMP-GEFII) to Rap1 signalling, which promotes insulin granule exocytosis. For SUs to exert their full effects in insulin secretion, they are required to activate Epac2 as well as to inhibit the β-cell K(ATP) channels. As Epac2 is also necessary for potentiation of glucose-induced insulin secretion by cAMP-increasing agents, such as incretin, Epac2 is a target of both cAMP and SUs. The distinct effects of various SUs appear to be because of their different actions on Epac2/Rap1 signalling as well as K(ATP) channels. Differently from other SUs, gliclazide is unique in that it is specific for β-cell K(ATP) channel and does not activate Epac2.
Collapse
Affiliation(s)
- S Seino
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan.
| | | | | | | |
Collapse
|
28
|
Gliclazide may have an antiapoptotic effect related to its antioxidant properties in human normal and cancer cells. Mol Biol Rep 2011; 39:5253-67. [PMID: 22183301 PMCID: PMC3310990 DOI: 10.1007/s11033-011-1323-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Accepted: 12/03/2011] [Indexed: 11/29/2022]
Abstract
Experimental and clinical studies suggest that gliclazide may protect pancreatic β-cells from apoptosis induced by an oxidative stress. However, the precise mechanism(s) of this action are not fully understood and requires further clarification. Therefore, using human normal and cancer cells we examined whether the anti-apoptotic effects of this sulfonylurea is due to its free radical scavenger properties. Hydrogen peroxide (H2O2) as a model trigger of oxidative stress was used to induce cell death. Our experiments were performed on human normal cell line (human umbilical vein endothelial cell line, HUVEC-c) and human cancer cell lines (human mammary gland cell line, Hs578T; human pancreatic duct epithelioid carcinoma cell line, PANC-1). To assess the effect of gliclazide the cells were pre-treated with the drug. The 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide assay was employed to measure the impact of gliclazide on cell viability. Generation of reactive oxygen species, mitochondrial membrane potential (∆Ψm), and intracellular Ca2+ concentration [Ca2+] were monitored. Furthermore, the morphological changes associated with apoptosis were determined using double staining with Hoechst 33258-propidium iodide (PI). Gliclazide protects the tested cells from H2O2-induced cell death most likely throughout the inhibition of ROS production. Moreover, the drug restored loss of ΔΨm and diminished intracellular [Ca2+] evoked by H2O2. Double staining with Hoechst 33258-PI revealed that pre-treatment with gliclazide diminished the number of apoptotic cells. Our findings indicate that gliclazide may protect both normal and cancer human cells against apoptosis induced by H2O2. It appears that the anti-apoptotic effect of the drug is most likely associated with reduction of oxidative stress.
Collapse
|
29
|
Lee JH, Lee JS, Kim YR, Jung WC, Lee KE, Lee SY, Hong EK. Hispidin Isolated from Phellinus linteus Protects Against Hydrogen Peroxide–Induced Oxidative Stress in Pancreatic MIN6N β-Cells. J Med Food 2011; 14:1431-8. [DOI: 10.1089/jmf.2010.1493] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Affiliation(s)
- Jung Hyun Lee
- Department of Bioengineering and Technology, Kangwon National University, Chuncheon, Korea
| | - Jong Seok Lee
- Department of Bioengineering and Technology, Kangwon National University, Chuncheon, Korea
| | - Young Rae Kim
- Department of Bioengineering and Technology, Kangwon National University, Chuncheon, Korea
| | - Woo Chul Jung
- Department of Bioengineering and Technology, Kangwon National University, Chuncheon, Korea
| | - Keun Eok Lee
- Department of Bioengineering and Technology, Kangwon National University, Chuncheon, Korea
| | - Shin Young Lee
- Department of Bioengineering and Technology, Kangwon National University, Chuncheon, Korea
| | - Eock Kee Hong
- Department of Bioengineering and Technology, Kangwon National University, Chuncheon, Korea
| |
Collapse
|
30
|
Lin CC, Raza A, Shih H. PEG hydrogels formed by thiol-ene photo-click chemistry and their effect on the formation and recovery of insulin-secreting cell spheroids. Biomaterials 2011; 32:9685-95. [PMID: 21924490 DOI: 10.1016/j.biomaterials.2011.08.083] [Citation(s) in RCA: 195] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 08/31/2011] [Indexed: 02/06/2023]
Abstract
Hydrogels provide three-dimensional frameworks with tissue-like elasticity and high permeability for culturing therapeutically relevant cells or tissues. While recent research efforts have created diverse macromer chemistry to form hydrogels, the mechanisms of hydrogel polymerization for in situ cell encapsulation remain limited. Hydrogels prepared from chain-growth photopolymerization of poly(ethylene glycol) diacrylate (PEGDA) are commonly used to encapsulate cells. However, free radical associated cell damage poses significant limitation for this gel platform. More recently, PEG hydrogels formed by thiol-ene photo-click chemistry have been developed for cell encapsulation. While both chain-growth and step-growth photopolymerizations offer spatial-temporal control over polymerization kinetics, step-growth thiol-ene hydrogels offer more diverse and preferential properties. Here, we report the superior properties of step-growth thiol-ene click hydrogels, including cytocompatibility of the reactions, improved hydrogel physical properties, and the ability for 3D culture of pancreatic β-cells. Cells encapsulated in thiol-ene hydrogels formed spherical clusters naturally and were retrieved via rapid chymotrypsin-mediated gel erosion. The recovered cell spheroids released insulin in response to glucose treatment, demonstrating the cytocompatibility of thiol-ene hydrogels and the enzymatic mechanism of cell spheroids recovery. Thiol-ene click reactions provide an attractive means to fabricate PEG hydrogels with superior gel properties for in situ cell encapsulation, as well as to generate and recover 3D cellular structures for regenerative medicine applications.
Collapse
Affiliation(s)
- Chien-Chi Lin
- Department of Biomedical Engineering, Purdue School of Engineering and Technology, Indiana University-Purdue University at Indianapolis, IN 46202, USA.
| | | | | |
Collapse
|
31
|
Abstract
Increasing evidences have suggested that oxidative stress plays a major role in the pathogenesis of diabetes mellitus (DM). Oxidative stress also appears to be the pathogenic factor in underlying diabetic complications. Reactive oxygen species (ROS) are generated by environmental factors, such as ionizing radiation and chemical carcinogens, and also by endogenous processes, including energy metabolism in mitochondria. ROS produced either endogenously or exogenously can attack lipids, proteins and nucleic acids simultaneously in living cells. There are many potential mechanisms whereby excess glucose metabolites traveling along these pathways might promote the development of DM complication and cause pancreatic β cell damage. However, all these pathways have in common the formation of ROS, that, in excess and over time, causes chronic oxidative stress, which in turn causes defective insulin gene expression and insulin secretion as well as increased apoptosis. Various methods for determining biomarkers of cellular oxidative stress have been developed, and some have been proposed for sensitive assessment of antioxidant defense and oxidative damage in diabetes and its complications. However, their clinical utility is limited by less than optimal standardization techniques and the lack of sufficient large-sized, multi-marker prospective trials.
Collapse
Affiliation(s)
- Hui Yang
- Department of Laboratory Medicine, China-Japan Friendship Hospital, Ministry of Health, Beijing, PR China
| | | | | | | |
Collapse
|
32
|
Palsamy P, Subramanian S. Ameliorative potential of resveratrol on proinflammatory cytokines, hyperglycemia mediated oxidative stress, and pancreatic beta-cell dysfunction in streptozotocin-nicotinamide-induced diabetic rats. J Cell Physiol 2010; 224:423-32. [PMID: 20333650 DOI: 10.1002/jcp.22138] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Chronic exposure of pancreatic beta-cells to supraphysiologic glucose causes adverse beta-cell dysfunction. Thus, the present study was aimed to investigate the hypothesis that oral administration of resveratrol attenuates hyperglycemia, proinflammatory cytokines and antioxidant competence and protects beta-cell ultrastructure in streptozotocin-nicotinamide-induced diabetic rats. Oral administration of resveratrol (5 mg/kg body weight) to diabetic rats for 30 days showed a significant decline in the levels of blood glucose, glycosylated hemoglobin (HbA1c), TNF-alpha, IL-1beta, IL-6, NF-kappaB p65 unit and nitric oxide (NO) with concomitant elevation in plasma insulin. Further, resveratrol treated diabetic rats elicited a notable attenuation in the levels of lipid peroxides, hydroperoxides and protein carbonyls in both plasma and pancreatic tissues. The diminished activities of pancreatic superoxide dismutase (SOD), catalase, glutathione peroxidase (GPx) and glutathione-S-transferase (GST) as well as the decreased levels of plasma ceruloplasmin, vitamin C, vitamin E and reduced glutathione (GSH) in diabetic rats were reverted to near normalcy by resveratrol administration. Based on histological and ultrastructural observations, it is first-time reported that the oral administration of resveratrol may effectively rescue beta-cells from oxidative damage without affecting their function and structural integrity. The results of the present investigation demonstrated that resveratrol exhibits significant antidiabetic potential by attenuating hyperglycemia, enhancing insulin secretion and antioxidant competence in pancreatic beta-cells of diabetic rats.
Collapse
Affiliation(s)
- P Palsamy
- Department of Biochemistry, University of Madras, Guindy Campus, Chennai, Tamilnadu, India
| | | |
Collapse
|
33
|
Pitocco D, Zaccardi F, Di Stasio E, Romitelli F, Santini SA, Zuppi C, Ghirlanda G. Oxidative stress, nitric oxide, and diabetes. Rev Diabet Stud 2010; 7:15-25. [PMID: 20703435 DOI: 10.1900/rds.2010.7.15] [Citation(s) in RCA: 190] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In the recent decades, oxidative stress has become focus of interest in most biomedical disciplines and many types of clinical research. Increasing evidence from research on several diseases show that oxidative stress is associated with the pathogenesis of diabetes, obesity, cancer, ageing, inflammation, neurodegenerative disorders, hypertension, apoptosis, cardiovascular diseases, and heart failure. Based on this research, the emerging concept is that oxidative stress is the "final common pathway", through which risk factors of several diseases exert their deleterious effects. Oxidative stress causes a complex dysregulation of cell metabolism and cell-cell homeostasis. In this review, we discuss the role of oxidative stress in the pathogenesis of insulin resistance and beta-cell dysfunction. These are the two most relevant mechanisms in the pathophysiology of type 2 diabetes, and in the pathogenesis of diabetic vascular complications, the leading cause of death in diabetic patients.
Collapse
Affiliation(s)
- Dario Pitocco
- Institute of Internal Medicine, Catholic University of Rome, Largo Agostino Gemelli 8, 00168 Rome, Italy.
| | | | | | | | | | | | | |
Collapse
|
34
|
Koh G, Lee DH, Woo JT. 2-Deoxy-D-ribose induces cellular damage by increasing oxidative stress and protein glycation in a pancreatic beta-cell line. Metabolism 2010; 59:325-32. [PMID: 19793592 DOI: 10.1016/j.metabol.2009.07.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Revised: 06/20/2009] [Accepted: 07/29/2009] [Indexed: 02/02/2023]
Abstract
2-Deoxy-D-ribose (dRib) is a sugar with a high reducing capacity. We previously reported that dRib induced damage in pancreatic beta-cells. The aim of this study was to investigate the mechanism of dRib-induced beta-cell damage. 2-Deoxy-D-ribose provoked cytotoxicity and apoptosis within 24 hours in HIT-T15 cells. Three antiglycating agents-diethylenetriaminepentaacetic acid, aminoguanidine, and pyridoxamine-dose dependently inhibited dRib-triggered cytotoxicity and significantly suppressed apoptosis induced by dRib. 2-Deoxy-d-ribose increased intracellular reactive oxygen species and protein carbonyl levels in a dose-dependent manner. Diethylenetriaminepentaacetic acid and aminoguanidine significantly reduced dRib-induced rises in intracellular reactive oxygen species. All 3 inhibitors decreased the production of intracellular protein carbonyls by dRib. On incubation with albumin, dRib increased dicarbonyl and advanced glycation end product formation. Aminoguanidine and pyridoxamine significantly decreased the dicarbonyl and advanced glycation end product augmentations. These results suggest that both oxidative stress and protein glycation are important mechanisms of dRib-induced damage in a pancreatic beta-cell line.
Collapse
Affiliation(s)
- Gwanpyo Koh
- Department of Internal Medicine, Jeju National University School of Medicine, Jeju 690-756, Republic of Korea.
| | | | | |
Collapse
|
35
|
Balasubramanian R, Ruiz de Azua I, Wess J, Jacobson KA. Activation of distinct P2Y receptor subtypes stimulates insulin secretion in MIN6 mouse pancreatic beta cells. Biochem Pharmacol 2010; 79:1317-26. [PMID: 20067775 DOI: 10.1016/j.bcp.2009.12.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 12/18/2009] [Accepted: 12/29/2009] [Indexed: 01/15/2023]
Abstract
Extracellular nucleotides and their receptor antagonists have therapeutic potential in disorders such as inflammation, brain disorders, and cardiovascular diseases. Pancreatic beta cells express several purinergic receptors, and reported nucleotide effects on insulin secretion are contradictory. We studied the effect of P2Y receptors on insulin secretion and cell death in MIN6, mouse pancreatic beta cells. Expression of P2Y(1) and P2Y(6) receptors was revealed by total mRNA analysis using RT-PCR. MIN6 cells were stimulated in the presence of 16.7 mM glucose with or without P2Y(1) and P2Y(6) agonists, 2-MeSADP and Up(3)U, respectively. Both the agonists increased insulin secretion with EC(50) values of 44.6+/-7.0 nM and 30.7+/-12.7 nM respectively. The insulin secretion by P2Y(1) and P2Y(6) agonists was blocked by their selective antagonists MRS2179 and MRS2578, respectively. Binding of the selective P2Y(1) receptor antagonist radioligand [125I]MRS2500 in MIN6 cell membranes was saturable (K(D) 4.74+/-0.47 nM), and known P2Y(1) ligands competed with high affinities. Inflammation and glucose toxicity lead to pancreatic beta cell death in diabetes. Flow cytometric analysis revealed that Up(3)U but not 2-MeSADP protected MIN6 cells against TNF-alpha induced apoptosis. Overall, the results demonstrate that selective stimulation of P2Y(1) and P2Y(6) receptors increases insulin secretion that accompanies intracellular calcium release, suggesting potential application of P2Y receptor ligands in the treatment of diabetes.
Collapse
Affiliation(s)
- Ramachandran Balasubramanian
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bldg. 8A, Rm. B1A-19, Bethesda, MD, USA
| | | | | | | |
Collapse
|
36
|
Gier B, Krippeit-Drews P, Sheiko T, Aguilar-Bryan L, Bryan J, Düfer M, Drews G. Suppression of KATP channel activity protects murine pancreatic beta cells against oxidative stress. J Clin Invest 2009; 119:3246-56. [PMID: 19805912 DOI: 10.1172/jci38817] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2009] [Accepted: 07/29/2009] [Indexed: 12/21/2022] Open
Abstract
The enhanced oxidative stress associated with type 2 diabetes mellitus contributes to disease pathogenesis. We previously identified plasma membrane-associated ATP-sensitive K+ (KATP) channels of pancreatic beta cells as targets for oxidants. Here, we examined the effects of genetic and pharmacologic ablation of KATP channels on loss of mouse beta cell function and viability following oxidative stress. Using mice lacking the sulfonylurea receptor type 1 (Sur1) subunit of KATP channels, we found that, compared with insulin secretion by WT islets, insulin secretion by Sur1-/- islets was less susceptible to oxidative stress induced by the oxidant H2O2. This was likely, at least in part, a result of the reduced ability of H2O2 to hyperpolarize plasma membrane potential and reduce cytosolic free Ca2+ concentration ([Ca2+]c) in the Sur1-/- beta cells. Remarkably, Sur1-/- beta cells were less prone to apoptosis induced by H2O2 or an NO donor than WT beta cells, despite an enhanced basal rate of apoptosis. This protective effect was attributed to upregulation of the antioxidant enzymes SOD, glutathione peroxidase, and catalase. Upregulation of antioxidant enzymes and reduced sensitivity of Sur1-/- cells to H2O2-induced apoptosis were mimicked by treatment with the sulfonylureas tolbutamide and gliclazide. Enzyme upregulation and protection against oxidant-induced apoptosis were abrogated by agents lowering [Ca2+]c. Sur1-/- mice were less susceptible than WT mice to streptozotocin-induced beta cell destruction and subsequent hyperglycemia and death, which suggests that loss of KATP channel activity may protect against streptozotocin-induced diabetes in vivo.
Collapse
Affiliation(s)
- Belinda Gier
- Institute of Pharmacy, Department of Pharmacology, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|
37
|
Zhao R, Li QW, Li J, Zhang T. Protective effect of Lycium barbarum polysaccharide 4 on kidneys in streptozotocin-induced diabetic rats. Can J Physiol Pharmacol 2009; 87:711-9. [DOI: 10.1139/y09-068] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Lycium barbarum polysaccharide (LBP) has been shown to have hypoglycemic and antioxidative properties, although its mode of action is yet unknown. Because oxidative stress is implicated in the pathogenesis of diabetic nephropathy, we evaluated the protective effect of LBP-4, the major active component of Lycium barbarum, on the defensive antioxidative mechanism in kidneys in a streptozotocin-induced diabetic rat model. Moreover, we investigated the effects of LBP-4 on the activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2) in isolated mesangial cells. The role of protein kinase C (PKC)-dependent and -independent pathways in LBP-4-reduced ERK1/2 was studied by bisindolylmaleimide (BIM) IV, an inhibitor of PKC. Diabetic rats treated with LBP-4 (10 mg/kg) for 8 weeks showed increased activity of antioxidant enzymes and increased scavenging of oxygen radicals, while the activity of PKC in the renal cortex was maintained at a physiological level. The decreased activation of ERK1/2 in mesangial cells, through the involvement of PKC, could explain the protective mechanism in kidneys of diabetic rats treated with LBP-4.
Collapse
Affiliation(s)
- Rui Zhao
- Department of Pharmaceutical Engineering, College of Life Science and Biotechnology, Heilongjiang August First Land Reclamation University, Daqing High-Tech Industrial Development Zone, 163319, P. R. China
- School of Animal Science, Northwest A & F University, 22 Xinong Street, Yangling 712100, P. R. China
- Department of Biological Engineering, School of Environment and Chemistry Engineering, Yanshan University, 438 Hebei Street, Qinhuangdao 066004, P. R. China
- School of Basic Medical Sciences, Jiamusi University, Jiamusi City, Heilongjiang Province, 154007, P. R. China
| | - Qing-wang Li
- Department of Pharmaceutical Engineering, College of Life Science and Biotechnology, Heilongjiang August First Land Reclamation University, Daqing High-Tech Industrial Development Zone, 163319, P. R. China
- School of Animal Science, Northwest A & F University, 22 Xinong Street, Yangling 712100, P. R. China
- Department of Biological Engineering, School of Environment and Chemistry Engineering, Yanshan University, 438 Hebei Street, Qinhuangdao 066004, P. R. China
- School of Basic Medical Sciences, Jiamusi University, Jiamusi City, Heilongjiang Province, 154007, P. R. China
| | - Jian Li
- Department of Pharmaceutical Engineering, College of Life Science and Biotechnology, Heilongjiang August First Land Reclamation University, Daqing High-Tech Industrial Development Zone, 163319, P. R. China
- School of Animal Science, Northwest A & F University, 22 Xinong Street, Yangling 712100, P. R. China
- Department of Biological Engineering, School of Environment and Chemistry Engineering, Yanshan University, 438 Hebei Street, Qinhuangdao 066004, P. R. China
- School of Basic Medical Sciences, Jiamusi University, Jiamusi City, Heilongjiang Province, 154007, P. R. China
| | - Tao Zhang
- Department of Pharmaceutical Engineering, College of Life Science and Biotechnology, Heilongjiang August First Land Reclamation University, Daqing High-Tech Industrial Development Zone, 163319, P. R. China
- School of Animal Science, Northwest A & F University, 22 Xinong Street, Yangling 712100, P. R. China
- Department of Biological Engineering, School of Environment and Chemistry Engineering, Yanshan University, 438 Hebei Street, Qinhuangdao 066004, P. R. China
- School of Basic Medical Sciences, Jiamusi University, Jiamusi City, Heilongjiang Province, 154007, P. R. China
| |
Collapse
|
38
|
Horsdal HT, Johnsen SP, Søndergaard F, Jacobsen J, Thomsen RW, Schmitz O, Sørensen HT, Rungby J. Sulfonylureas and prognosis after myocardial infarction in patients with diabetes: a population-based follow-up study. Diabetes Metab Res Rev 2009; 25:515-22. [PMID: 19459168 DOI: 10.1002/dmrr.971] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND The cardiovascular safety, including risk of myocardial infarction (MI), of individual sulfonylureas (SUs) may differ. It remains uncertain whether treatment with individual SUs influences prognosis following MI. METHODS We conducted a nationwide population-based follow-up study among all Danish patients hospitalized with first-time MI from 1996 to 2004. From the national health databases, we identified 3930 MI patients who used SUs at the time of admission. We computed mortality rates and rates of MI and heart failure readmission according to type of SU and used Cox's proportional hazards regression analysis to compute hazard ratios (HRs) as estimates of relative risk controlling for differences in prognostic covariates. RESULTS The 30-day and 1-year mortality after MI among SU users was 22.0% and 35.3%, respectively. We found no substantial differences in 30-day and 1-year mortality among users of different SUs. Use of gliclazide in monotherapy showed a trend towards lower mortality; adjusted HR of 1-year mortality 0.70 (95% CI: 0.48-1.00). Users of the different SUs appeared to have similar risks of new MI and heart failure following MI. CONCLUSIONS The prognosis after MI was not substantially influenced by the choice of SU.
Collapse
Affiliation(s)
- Henriette T Horsdal
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Del Guerra S, D’Aleo V, Lupi R, Masini M, Bugliani M, Boggi U, Filipponi F, Marchetti P. Effects of exposure of human islet beta-cells to normal and high glucose levels with or without gliclazide or glibenclamide. DIABETES & METABOLISM 2009; 35:293-8. [DOI: 10.1016/j.diabet.2009.01.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2008] [Revised: 01/02/2009] [Accepted: 01/06/2009] [Indexed: 11/15/2022]
|
40
|
Kinukawa J, Shimura M, Harata N, Tamai M. Gliclazide Attenuates the Intracellular Ca2+Changes InducedIn Vitroby Ischemia in the Retinal Slices of Rats with Streptozotocin-Induced Diabetes. Curr Eye Res 2009; 30:789-98. [PMID: 16146925 DOI: 10.1080/02713680591002808] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE To investigate the dynamics of the intracellular Ca2+ concentration ([Ca2+]i) during retinal ischemia in rats with streptozotocin (STZ)-induced diabetes and the effect of gliclazide, a sulfonylurea with a potent free-radical scavenging activity on ischemia-induced [Ca2+]i dynamics. METHODS Rats with STZ (65 mg/kg) induced diabetes were divided into three groups: the untreated diabetic group, the gliclazide-treated group, and the glibenclamide-treated group. An ischemic condition was imposed in vitro on the retinal slices by perfusion with an oxygen/glucose deprived solution. The [Ca2+]i was measured in individual layers of the rat retinal slices loaded with the Ca2+ indicator fluo-3. RESULTS As compared to that in the normal rat retina, both the amplitude and the kinetics of the [Ca2+]i increase were suppressed in the intermediate layers of the retinal slices from the diabetic rats under the ischemic condition. These changes were attenuated by the administration of gliclazide but not by that of glibenclamide. CONCLUSIONS Hyperglycemia influences ischemia-induced [Ca2+]i dynamics predominantly in the intermediate layers of the retina, and gliclazide, as compared to glibenclamide without a free radical scavenging activity, potently attenuates the ischemia-induced changes in the calcium dynamics.
Collapse
Affiliation(s)
- Jiro Kinukawa
- Department of Ophthalmology and Medical Science, Tohoku University Graduate School of Medicine, Sendai Miyagi, Japan
| | | | | | | |
Collapse
|
41
|
Ramkumar KM, Manjula C, Sankar L, Suriyanarayanan S, Rajaguru P. Potential in vitro antioxidant and protective effects of Gymnema montanum H. on alloxan-induced oxidative damage in pancreatic beta-cells, HIT-T15. Food Chem Toxicol 2009; 47:2246-56. [PMID: 19520139 DOI: 10.1016/j.fct.2009.06.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2009] [Revised: 05/24/2009] [Accepted: 06/04/2009] [Indexed: 11/15/2022]
Abstract
The present study describes the antioxidant activities of ethanol extract from Gymnema montanum (GLEt) which is an endemic plant of India. Antioxidant activity of the GLEt was studied in vitro based on scavenging of hydroxyl radicals, superoxide anions, nitric oxide, hydrogen peroxide, peroxynitrite, reducing power and inhibition of lipid peroxidation estimated in terms of thiobarbituric acid reactive substances (TBARS). Further, we examined its protective effect against alloxan-induced oxidative stress in pancreatic beta-cells, HIT-T15 by measuring the free radical generation, malonaldehyde formation and antioxidant levels such as CAT, GPx and GSH. Results showed that G. montanum leaves exhibited significant antioxidant activities measured by various in vitro model systems. The HIT-T15 cell line studies showed the tendency of GLEt to increase antioxidant levels meanwhile decrease the free radical formation and inhibit the lipid peroxidation. The antioxidant activity was found to be well correlated with the phenolic phytochemicals present in the extract. GC-MS analyses revealed the presence of few phenolic compounds in the extract. As this plant has already been demonstrated for a variety of medicinal properties from our laboratory, results of this study suggest that G. montanum is an interesting source for antioxidant compounds and useful for various therapeutic applications.
Collapse
Affiliation(s)
- Kunga Mohan Ramkumar
- Department of Biotechnology, Anna University-Tiruchirappalli, Tamil Nadu, India.
| | | | | | | | | |
Collapse
|
42
|
Abstract
We investigated the influence of short-term culture in vitro on the appearance of apoptosis of human fetal pancreatic islets (HFIs) and its effect on the mass and insulin-secretory capacity (ISC) of ?-cells. It was found that apoptosis was present from the end of the culture period, increasing as a function of time and leading to decrease of ?-cell mass. At the same time, ISC decreased. The decrease of ?-cell mass and ISC may influence significantly the clinical outcome of HFIs transplantation in type 1 diabetic patients.
Collapse
|
43
|
Sawada F, Inoguchi T, Tsubouchi H, Sasaki S, Fujii M, Maeda Y, Morinaga H, Nomura M, Kobayashi K, Takayanagi R. Differential effect of sulfonylureas on production of reactive oxygen species and apoptosis in cultured pancreatic beta-cell line, MIN6. Metabolism 2008; 57:1038-45. [PMID: 18640379 DOI: 10.1016/j.metabol.2008.01.038] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Accepted: 01/10/2008] [Indexed: 01/09/2023]
Abstract
Sulfonylureas are considered to cause beta-cell apoptosis. However, it is unclear how this occurs and whether there is a difference in such effects among various sulfonylureas. Here, we examined the effects of various sulfonylureas and a short-acting insulin secretagogue, nateglinide, on oxidative stress and apoptosis using the beta-cell line MIN6. After cultured MIN6 cells were exposed to various concentrations of sulfonylureas (glibenclamide, glimepiride, and gliclazide) or nateglinide, intracellular production of reactive oxygen species (ROS) was evaluated by staining with 2',7'-dichlorofluorescein diacetate. The effect of these agents on apoptosis was also evaluated by the terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-biotin nick-end labeling technique. Exposure of beta-cells to glibenclamide, glimepiride, and nateglinide significantly increased intracellular ROS production in a concentration-dependent manner (0.1-10 micromol/L). These effects were completely blocked by nicotinamide adenine dinucleotide phosphate [NAD(P)H] oxidase inhibitors (diphenylene iodonium or apocynin) or a protein kinase C inhibitor (calphostin C). After exposure to these agents for 48 hours, the numbers of apoptotic cells were also significantly increased. These effects were significantly blocked by apocynin and antioxidant N-acetyl-L-cysteine. In contrast, exposure to any concentrations of gliclazide did not affect either intracellular ROS production or the numbers of apoptotic cells. Sulfonylureas (glibenclamide and glimepiride, but not gliclazide) and nateglinide stimulated ROS production via protein kinase C-dependent activation of NAD(P)H oxidase and consequently caused beta-cell apoptosis in vitro. Because of the lack of such adverse effects, gliclazide may have a benefit in the preservation of functional beta-cell mass.
Collapse
Affiliation(s)
- Fumi Sawada
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Glucotoxicity, lipotoxicity, and glucolipotoxicity are secondary phenomena that are proposed to play a role in all forms of type 2 diabetes. The underlying concept is that once the primary pathogenesis of diabetes is established, probably involving both genetic and environmental forces, hyperglycemia and very commonly hyperlipidemia ensue and thereafter exert additional damaging or toxic effects on the beta-cell. In addition to their contribution to the deterioration of beta-cell function after the onset of the disease, elevations of plasma fatty acid levels that often accompany insulin resistance may, as glucose levels begin to rise outside of the normal range, also play a pathogenic role in the early stages of the disease. Because hyperglycemia is a prerequisite for lipotoxicity to occur, the term glucolipotoxicity, rather than lipotoxicity, is more appropriate to describe deleterious effects of lipids on beta-cell function. In vitro and in vivo evidence supporting the concept of glucotoxicity is presented first, as well as a description of the underlying mechanisms with an emphasis on the role of oxidative stress. Second, we discuss the functional manifestations of glucolipotoxicity on insulin secretion, insulin gene expression, and beta-cell death, and the role of glucose in the mechanisms of glucolipotoxicity. Finally, we attempt to define the role of these phenomena in the natural history of beta-cell compensation, decompensation, and failure during the course of type 2 diabetes.
Collapse
Affiliation(s)
- Vincent Poitout
- Montreal Diabetes Research Center, CR-CHUM, Technopole Angus, 2901 Rachel Est, Montreal, Quebec, Canada H1W 4A4.
| | | |
Collapse
|
45
|
Corgnali M, Piconi L, Ihnat M, Ceriello A. Evaluation of gliclazide ability to attenuate the hyperglycaemic 'memory' induced by high glucose in isolated human endothelial cells. Diabetes Metab Res Rev 2008; 24:301-9. [PMID: 18088078 DOI: 10.1002/dmrr.804] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Patients with long-term exposure to high levels of hyperglycaemia remain more susceptible to diabetes-related complications, even with subsequent lower levels of hyperglycaemia. We sought to confirm the hypothesis that exposure to continuous increased glucose results in a memory of cellular stress in isolated endothelial cells, even when switched back to normal glucose, and to investigate the ability of gliclazide to attenuate this phenomenon. METHODS Human umbilical vein endothelial cells were incubated for 21 days in normal glucose (5 mmol/L), high glucose (30 mmol/L), or high glucose for 14 days followed by normal glucose for 7 days (memory condition). The effects of gliclazide (10 micromol/L) and glibenclamide (1 micromol/L) were evaluated in the memory condition and added to the culture media early (first 14 days), late (last 7 days), or throughout the study. Oxidative stress and cell apoptosis parameters were investigated. RESULTS Continuous high glucose increased reactive oxygen species, 8-OHdG, nitrotyrosine, caspase-3, and reduced Bcl-2 expression. These deleterious effects were also observed in the memory condition. Gliclazide applied early or throughout the study improved all parameters. In contrast, glibenclamide showed no relevant effect on study parameters. CONCLUSIONS Our results suggest that gliclazide prevents endothelial cell apoptosis by reducing oxidative stress. The results appear to confirm the hypothesis that exposure of cells to continuous increased glucose results in a hyperglycaemic cellular memory that remains, even when cells are switched back to normal glucose. Gliclazide attenuated this cellular memory, decreasing oxidative stress and protecting vascular endothelial cells from apoptosis.
Collapse
|
46
|
In vitro effect of gliclazide on DNA damage and repair in patients with type 2 diabetes mellitus (T2DM). Chem Biol Interact 2008; 173:159-65. [PMID: 18485337 DOI: 10.1016/j.cbi.2008.03.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Revised: 03/27/2008] [Accepted: 03/28/2008] [Indexed: 11/20/2022]
Abstract
Type 2 diabetes mellitus is associated with elevated level of oxidative stress, which is one of the most important factors responsible for the development of chronic complications of this disease. Moreover, it was shown that diabetic patients had increased level of oxidative DNA damage and decreased effectiveness of DNA repair. These changes may be associated with increased risk of cancer in T2DM patients, since DNA damage and DNA repair play a pivotal role in malignant transformation. It was found that gliclazide, an oral hypoglycemic drug with antioxidant properties, diminished DNA damage induced by free radicals. Therefore, the aim of the present study was to evaluate the in vitro impact of gliclazide on: (i) endogenous basal and oxidative DNA damage, (ii) DNA damage induced by hydrogen peroxide and (iii) the efficacy of DNA repair of such damage. DNA damage and DNA repair in peripheral blood lymphocytes of 30 T2DM patients and 30 non-diabetic individuals were evaluated by alkaline single cell electrophoresis (comet) assay. The extent of oxidative DNA damage was assessed by DNA repair enzymes: endonuclease III and formamidopyrimidine-DNA glycosylase. The endogenous basal and oxidative DNA damages were higher in lymphocytes of T2DM patients compared to non-diabetic subjects and gliclazide decreased the level of such damage. The drug significantly decreased the level of DNA damage induced by hydrogen peroxide in both groups. Gliclazide increased the effectiveness of DNA repair in lymphocytes of T2DM patients (93.4% (with gliclazide) vs 79.9% (without gliclazide); P< or =0.001) and non-diabetic subjects (95.1% (with gliclazide) vs 90.5% (without gliclazide); P< or =0.001). These results suggest that gliclazide may protect against the oxidative stress-related chronic diabetes complications, including cancer, by decreasing the level of DNA damage induced by reactive oxygen species.
Collapse
|
47
|
Dixit PP, Devasagayam TP, Ghaskadbi S. Formulated antidiabetic preparation Syndrex® has a strong antioxidant activity. Eur J Pharmacol 2008; 581:216-25. [DOI: 10.1016/j.ejphar.2007.11.052] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2007] [Revised: 11/20/2007] [Accepted: 11/21/2007] [Indexed: 01/25/2023]
|
48
|
Standl E, Schnell O. Insulin as a first-line therapy in type 2 diabetes: should the use of sulfonylureas be halted? Diabetes Care 2008; 31 Suppl 2:S136-9. [PMID: 18227474 DOI: 10.2337/dc08-s234] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Eberhard Standl
- Munich Diabetes Research Institute, Kölner Pl.1, D-80804 Munich, Germany.
| | | |
Collapse
|
49
|
Pallardo Sánchez L. Sulfonilureas en el tratamiento del paciente con diabetes mellitus tipo 2. ACTA ACUST UNITED AC 2008. [DOI: 10.1016/s1575-0922(08)76259-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
50
|
Bonora E. Protection of pancreatic beta-cells: is it feasible? Nutr Metab Cardiovasc Dis 2008; 18:74-83. [PMID: 18096375 DOI: 10.1016/j.numecd.2007.05.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2007] [Revised: 05/03/2007] [Accepted: 05/18/2007] [Indexed: 12/20/2022]
Abstract
Hyperglycemia, which is the biochemical hallmark of type 2 diabetes, mainly results from insulin resistance and beta-cell dysfunction. However, the latter is crucial in the development of the disease because diabetes cannot occur without an impairment of insulin secretion. Beta-cell failure is also responsible for progressive loss of metabolic control in type 2 diabetic patients and the eventual need for insulin treatment. An impairment of beta-cell function can be detected in several ways and can be observed already in pre-diabetic individuals. Histopathology studies documented that beta-cell volume is reduced in pre-diabetes and, to a greater extent, in type 2 diabetes mainly because the apoptotic rate of beta-cells is increased whereas neogenesis is intact. All anti-diabetic agents can improve, directly or indirectly, beta-cell function. However, only PPAR-gamma agonists and incretin-mimetic agents seem to have favorable effects on beta-cell morphology and volume. Many trials showed that type 2 diabetes can be prevented but few of them directly addressed the issue of beta-cell protection by the intervention used in the study. It is reasonable to conclude that in these trials diabetes prevention, which was based on the use of lifestyle changes (diet and/or exercise) or different drugs (tolbutamide, acarbose, metformin, glitazones, bezafibrate, orlistat, angiotensin converting enzyme inhibitors, angiotensin II receptor blockers or pravastatin), depended also, or mainly, on a protection of the beta-cells but in most studies data on insulin secretion are not available or are insufficient to draw firm conclusions. The mechanisms of beta-cell protection in these trials, if any, remain unknown. They could be various and likely included reduced glucotoxicity, lipotoxicity, insulin resistance, inflammation, oxidant stress and/or apoptosis, an amelioration of islet blood flow and/or favorable changes in cation balance within the islets. Contrasting the decline and the eventual failure of beta-cells is crucial in preventing type 2 diabetes as well as in changing the natural history of the disease, when it occurs. The protection can be achieved in several ways but any strategy should include a change in lifestyle in order to generate a healthier islet milieu. Among anti-diabetic drugs, PPAR-gamma agonists and incretin-mimetic agents are the most promising in the protection. Among other drugs, inhibitors of the renin-angiotensin system might play a significant role. The increased worldwide diffusion of type 2 diabetes and the progressive loss of metabolic control in affected patients are clear demonstrations that the strategies to protect the beta-cells implemented so far, if any, were largely inadequate. Anti-diabetic agents targeting the intimate mechanisms of beta-cell damage might change the scenario in the near future.
Collapse
Affiliation(s)
- Enzo Bonora
- Division of Endocrinology and Metabolic Diseases, Department of Biomedical and Surgical Sciences, University of Verona, Verona, Italy.
| |
Collapse
|