1
|
Holendová B, Benáková Š, Křivonosková M, Plecitá-Hlavatá L. Redox Status as a Key Driver of Healthy Pancreatic Beta-Cells. Physiol Res 2024; 73:S139-S152. [PMID: 38647167 PMCID: PMC11412338 DOI: 10.33549/physiolres.935259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Redox status plays a multifaceted role in the intricate physiology and pathology of pancreatic beta-cells, the pivotal regulators of glucose homeostasis through insulin secretion. They are highly responsive to changes in metabolic cues where reactive oxygen species are part of it, all arising from nutritional intake. These molecules not only serve as crucial signaling intermediates for insulin secretion but also participate in the nuanced heterogeneity observed within the beta-cell population. A central aspect of beta-cell redox biology revolves around the localized production of hydrogen peroxide and the activity of NADPH oxidases which are tightly regulated and serve diverse physiological functions. Pancreatic beta-cells possess a remarkable array of antioxidant defense mechanisms although considered relatively modest compared to other cell types, are efficient in preserving redox balance within the cellular milieu. This intrinsic antioxidant machinery operates in concert with redox-sensitive signaling pathways, forming an elaborate redox relay system essential for beta-cell function and adaptation to changing metabolic demands. Perturbations in redox homeostasis can lead to oxidative stress exacerbating insulin secretion defect being a hallmark of type 2 diabetes. Understanding the interplay between redox signaling, oxidative stress, and beta-cell dysfunction is paramount for developing effective therapeutic strategies aimed at preserving beta-cell health and function in individuals with type 2 diabetes. Thus, unraveling the intricate complexities of beta-cell redox biology presents exciting avenues for advancing our understanding and treatment of metabolic disorders.
Collapse
Affiliation(s)
- B Holendová
- Laboratory of Pancreatic Islet Research, Czech Academy of Sciences, Prague 4, Czech Republic.
| | | | | | | |
Collapse
|
2
|
Salama A, Elgohary R, Amin MM, Elwahab SA. Impact of protocatechuic acid on alleviation of pulmonary damage induced by cyclophosphamide targeting peroxisome proliferator activator receptor, silent information regulator type-1, and fork head box protein in rats. Inflammopharmacology 2023; 31:1361-1372. [PMID: 36877411 DOI: 10.1007/s10787-023-01156-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 02/06/2023] [Indexed: 03/07/2023]
Abstract
Cyclophosphamide (CP) is a chemotherapeutic agent that causes pulmonary damage by generating free radicals and pro-inflammatory cytokines. Pulmonary damage has a high mortality rate due to the severe inflammation and edema occurred in lung. PPARγ/Sirt 1 signaling has been shown to be cytoprotective effect against cellular inflammatory stress and oxidative injury. Protocatechuic acid (PCA) is a potent Sirt1 activator and exhibits antioxidant as well as anti-inflammatory properties. The current study aims to investigate the therapeutic impacts of PCA against CP-induced pulmonary damage in rats. Rats were assigned randomly into 4 experimental groups. The control group was injected with a single i.p injection of saline. CP group was injected with a single i.p injection of CP (200 mg/kg). PCA groups were administered orally with PCA (50 and 100 mg/kg; p.o.) once daily for 10 consecutive days after CP injection. PCA treatment resulted in a significant decrease in the protein levels of MDA, a marker of lipid peroxidation, NO and MPO along with a significant increase in GSH and catalase protein levels. Moreover, PCA downregulated anti-inflammatory markers as IL-17, NF-κB, IKBKB, COX-2, TNF-α, and PKC and upregulated cytoprotective defenses as PPARγ, and SIRT1. In addition, PCA administration ameliorated FoxO-1 elevation, increased Nrf2 gene expression, and reduced air alveoli emphysema, bronchiolar epithelium hyperplasia and inflammatory cell infiltration induced by CP. PCA might represent a promising adjuvant to prevent pulmonary damage in patients receiving CP due to its antioxidant and anti-inflammatory effects with cytoprotective defenses.
Collapse
Affiliation(s)
- Abeer Salama
- Pharmacology Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El Buhouth St. (Former El-Tahrir St.), Dokki, Cairo, 12622, Egypt
| | - Rania Elgohary
- Narcotics, Ergogenics and Poisons Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El Buhouth St. (Former El-Tahrir St.), Dokki, Cairo, 12622, Egypt.
| | - Mohamed M Amin
- Pharmacology Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El Buhouth St. (Former El-Tahrir St.), Dokki, Cairo, 12622, Egypt
| | - Sahar Abd Elwahab
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine Cairo University Al Kasr Al Aini, Old Cairo, Cairo Governorate, Egypt
| |
Collapse
|
3
|
Zmazek J, Grubelnik V, Markovič R, Marhl M. Modeling the Amino Acid Effect on Glucagon Secretion from Pancreatic Alpha Cells. Metabolites 2022; 12:metabo12040348. [PMID: 35448534 PMCID: PMC9028923 DOI: 10.3390/metabo12040348] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 11/29/2022] Open
Abstract
Type 2 Diabetes Mellitus (T2DM) is a burdensome problem in modern society, and intensive research is focused on better understanding the underlying cellular mechanisms of hormone secretion for blood glucose regulation. T2DM is a bi-hormonal disease, and in addition to 100 years of increasing knowledge about the importance of insulin, the second hormone glucagon, secreted by pancreatic alpha cells, is becoming increasingly important. We have developed a mathematical model for glucagon secretion that incorporates all major metabolic processes of glucose, fatty acids, and glutamine as the most abundant postprandial amino acid in blood. In addition, we consider cAMP signaling in alpha cells. The model predictions quantitatively estimate the relative importance of specific metabolic and signaling pathways and particularly emphasize the important role of glutamine in promoting glucagon secretion, which is in good agreement with known experimental data.
Collapse
Affiliation(s)
- Jan Zmazek
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia; (J.Z.); (R.M.)
| | - Vladimir Grubelnik
- Faculty of Electrical Engineering and Computer Science, University of Maribor, 2000 Maribor, Slovenia;
| | - Rene Markovič
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia; (J.Z.); (R.M.)
- Faculty of Electrical Engineering and Computer Science, University of Maribor, 2000 Maribor, Slovenia;
| | - Marko Marhl
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia; (J.Z.); (R.M.)
- Faculty of Education, University of Maribor, 2000 Maribor, Slovenia
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
- Correspondence:
| |
Collapse
|
4
|
Improving effect of cordycepin on insulin synthesis and secretion in normal and oxidative-damaged INS-1 cells. Eur J Pharmacol 2022; 920:174843. [DOI: 10.1016/j.ejphar.2022.174843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 02/05/2022] [Accepted: 02/16/2022] [Indexed: 01/18/2023]
|
5
|
Bacillus toyonensis SAU-19 Ameliorates Hepatic Insulin Resistance in High-Fat Diet/Streptozocin-Induced Diabetic Mice. Nutrients 2021; 13:nu13124512. [PMID: 34960064 PMCID: PMC8703646 DOI: 10.3390/nu13124512] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/15/2021] [Accepted: 12/15/2021] [Indexed: 12/16/2022] Open
Abstract
Insulin resistance (IR) is a hallmark of type 2 diabetes mellitus (T2DM). This study was performed to investigate the antidiabetic effect of Bacillus toyonensis SAU-19 and its possible mechanisms of action in mice with type 2 diabetes mellitus (T2DM). Thirty SPFKM mice were randomly assigned to three groups: control, diabetic model, and diabetes + Bacillus toyonensis SAU-19 group. After 35 days, blood was collected for biochemical analysis and liver tissue samples for histopathological analysis using H&E staining, qPCR, and ELISA. The results showed that the administration of B. toyonensis SAU-19 significantly improved the blood glucose, hepatic insulin resistance, and morphological changes of the liver characterized by significant improvement of dyslipidemia, glycogen synthesis, and antioxidant status (p < 0.05), indicating the strains’ ameliorating effects on hepatic insulin resistance in T2DM. In conclusion, the probiotic strain (B. toyonensis SAU-19) inhibits T2DM by reducing insulin resistance, improving antioxidant status, and downregulating genes related to glucose synthesis; hence, it may be used in treating diabetes and other metabolic disorders. This study provides the basis for further studies into the molecular mechanisms of B. toyonensis SAU-19 in treating T2DM.
Collapse
|
6
|
Song J, He Q, Guo X, Wang L, Wang J, Cui C, Hu H, Yang M, Cui Y, Zang N, Yan F, Liu F, Sun Y, Liang K, Qin J, Zhao R, Wang C, Sun Z, Hou X, Li W, Chen L. Mesenchymal stem cell-conditioned medium alleviates high fat-induced hyperglucagonemia via miR-181a-5p and its target PTEN/AKT signaling. Mol Cell Endocrinol 2021; 537:111445. [PMID: 34464683 DOI: 10.1016/j.mce.2021.111445] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/08/2021] [Accepted: 08/25/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND α-cell dysregulation gives rise to fasting and postprandial hyperglycemia in type 2 diabetes mellitus(T2DM). Administration of Mesenchymal stem cells (MSCs) or their conditioned medium can improve islet function and enhance insulin secretion. However, studies showing the direct effect of MSCs on islet α-cell dysfunction are limited. METHODS In this study, we used high-fat diet (HFD)-induced mice and α-cell line exposure to palmitate (PA) to determine the effects of bone marrow-derived MSC-conditioned medium (bmMSC-CM) on glucagon secretion. Plasma and supernatant glucagon were detected by enzyme-linked immunosorbent assay(ELISA). To investigate the potential signaling pathways, phosphatase and tensin homolog deleted on chromosome 10 (PTEN), AKT and phosphorylated AKT(p-AKT) were assessed by Western blotting. RESULTS In vivo, bmMSC-CM infusion improved the glucose and insulin tolerance and protected against HFD-induced hyperglycemia and hyperglucagonemia. Meanwhile, bmMSC-CM infusion ameliorated HFD-induced islet hypertrophy and decreased α- and β-cell area. Consistently, in vitro, glucagon secretion from α-cells or primary islets was inhibited by bmMSC-CM, accompanied by reduction of intracellular PTEN expression and restoration of AKT signaling. Previous studies and the TargetScan database indicate that miR-181a and its target PTEN play vital roles in ameliorating α-cell dysfunction. We observed that miR-181a-5p was highly expressed in BM-MSCs but prominently lower in αTC1-6 cells. Overexpression or downregulation of miR-181a-5p respectively alleviated or aggravated glucagon secretion in αTC1-6 cells via the PTEN/AKT signaling pathway. CONCLUSIONS Our observations suggest that MSC-derived miR-181a-5p mitigates glucagon secretion of α-cells by regulating PTEN/AKT signaling, which provides novel evidence demonstrating the potential for MSCs in treating T2DM.
Collapse
Affiliation(s)
- Jia Song
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Qin He
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Xinghong Guo
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Lingshu Wang
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Jinbang Wang
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Chen Cui
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Huiqing Hu
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Mengmeng Yang
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Yixin Cui
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Nan Zang
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Fei Yan
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Fuqiang Liu
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Yujing Sun
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Kai Liang
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Jun Qin
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Ruxing Zhao
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Chuan Wang
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Zheng Sun
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Xinguo Hou
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China; Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan, 250012, Shandong, China; Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine & Health, Jinan, 250012, Shandong, China; Jinan Clinical Research Center for Endocrine and Metabolic Disease, Jinan, 250012, Shandong, China
| | - Wenjuan Li
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China; Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan, 250012, Shandong, China; Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine & Health, Jinan, 250012, Shandong, China; Jinan Clinical Research Center for Endocrine and Metabolic Disease, Jinan, 250012, Shandong, China.
| | - Li Chen
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China; Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan, 250012, Shandong, China; Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine & Health, Jinan, 250012, Shandong, China; Jinan Clinical Research Center for Endocrine and Metabolic Disease, Jinan, 250012, Shandong, China.
| |
Collapse
|
7
|
Novelli M, Beffy P, Masini M, Vantaggiato C, Martino L, Marselli L, Marchetti P, De Tata V. Selective beta-cell toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin on isolated pancreatic islets. CHEMOSPHERE 2021; 265:129103. [PMID: 33288281 DOI: 10.1016/j.chemosphere.2020.129103] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 11/20/2020] [Accepted: 11/22/2020] [Indexed: 06/12/2023]
Abstract
An association between exposure to environmental pollutants and diabetes risk has been repeatedly shown by epidemiological studies. However, the biological basis of this association still need to be clarified. In this research we explored the effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure on isolated pancreatic islets. After 1, 6 and 24 h exposure of isolated islets to different concentrations (1-50 nM) of TCDD we assayed: i) cell survival; ii) ultrastructure; iii) glucose-stimulated insulin secretion (GSIS); iv) expression of selected genes. A significant, dose-related increase of both necrosis and apoptosis was observed isolated rat islets after 24 h exposure to TCDD. The electron microscopic analysis revealed, at the same time point, the presence of several ultrastructural alterations (mitochondrial swelling, increased mitophagy, dilation of the endoplasmic reticulum) that, very interestingly, were exclusively observed in beta cells and not in other endocrine cells. Similar results were obtained in isolated human islets. GSIS was rapidly (1 h) and persistently (6 and 24 h) decreased by TCDD exposure even at the smallest concentration (1 nM). TCDD exposure significantly affected gene expression in isolated islets: Glut2, Gck, Bcl-xL, MafA, Pdx1 FoxO1 and IRE1 gene expression was significantly decreased, whereas Puma, DP5, iNOS and Chop gene expression was significantly increased after 6 h exposure to TCDD. In conclusion, our results clearly indicated that pancreatic beta cells represent not only a sensitive but also a specific target of the toxic action of dioxin.
Collapse
Affiliation(s)
- Michela Novelli
- Department of Translational Research and New Technologies in Medicine and Surgery, Italy
| | - Pascale Beffy
- Department of Translational Research and New Technologies in Medicine and Surgery, Italy
| | - Matilde Masini
- Department of Translational Research and New Technologies in Medicine and Surgery, Italy
| | - Chiara Vantaggiato
- Department of Translational Research and New Technologies in Medicine and Surgery, Italy
| | - Luisa Martino
- Department of Translational Research and New Technologies in Medicine and Surgery, Italy
| | | | | | - Vincenzo De Tata
- Department of Translational Research and New Technologies in Medicine and Surgery, Italy; CIME (Centro Interdipartimentale di Microscopia Elettronica), University of Pisa, Pisa, Italy.
| |
Collapse
|
8
|
Mechanisms of the Regulation and Dysregulation of Glucagon Secretion. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3089139. [PMID: 32774668 PMCID: PMC7396046 DOI: 10.1155/2020/3089139] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 07/11/2020] [Indexed: 02/06/2023]
Abstract
Glucagon, a hormone secreted by pancreatic alpha cells, contributes to the maintenance of normal blood glucose concentration by inducing hepatic glucose production in response to declining blood glucose. However, glucagon hypersecretion contributes to the pathogenesis of type 2 diabetes. Moreover, diabetes is associated with relative glucagon undersecretion at low blood glucose and oversecretion at normal and high blood glucose. The mechanisms of such alpha cell dysfunctions are not well understood. This article reviews the genesis of alpha cell dysfunctions during the pathogenesis of type 2 diabetes and after the onset of type 1 and type 2 diabetes. It unravels a signaling pathway that contributes to glucose- or hydrogen peroxide-induced glucagon secretion, whose overstimulation contributes to glucagon dysregulation, partly through oxidative stress and reduced ATP synthesis. The signaling pathway involves phosphatidylinositol-3-kinase, protein kinase B, protein kinase C delta, non-receptor tyrosine kinase Src, and phospholipase C gamma-1. This knowledge will be useful in the design of new antidiabetic agents or regimens.
Collapse
|
9
|
Castex F, Leroy J, Broca C, Mezghenna K, Duranton F, Lavallard V, Lebreton F, Gross R, Wojtusciszyn A, Lajoix AD. Differential sensitivity of human islets from obese versus lean donors to chronic high glucose or palmitate. J Diabetes 2020; 12:532-541. [PMID: 32090456 DOI: 10.1111/1753-0407.13026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 01/15/2020] [Accepted: 02/19/2020] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Due to the shortage of multi-organ donors, human pancreatic islet transplantation has now been extended to islets originating from obese subjects. In this study, our aim is to compare the respective sensitivity of human islets from lean vs obese donors to chronic high glucose or high palmitate. METHODS Human islets were isolated from pancreases harvested from brain-dead multi-organ donors. Islets were cultured during 72 hours in the presence of moderate (16.7 mmol/L) or high (28 mmoL/L) glucose concentrations, or glucose (5.6 mmoL/L) and palmitate (0.4 mmoL/L), before measurement of their response to glucose. RESULTS We first observed a greater insulin response in islets from obese donors under both basal and high-glucose conditions, confirming their hyperresponsiveness to glucose. When islets from obese donors were cultured in the presence of moderate or high glucose concentrations, insulin response to glucose remained unchanged or was slightly reduced, as opposed to that observed in lean subjects. Moreover, culturing islets from obese donors with high palmitate also induced less reduction in insulin response to glucose than in lean subjects. This partial protection of obese islets is associated with less induction of inducible nitric oxide synthase in islets, together with a greater expression of the transcription factor forkhead box O1 (FOXO1). CONCLUSIONS Our data suggest that in addition to an increased sensitivity to glucose, islets from obese subjects can be considered as more resistant to glucose and fatty acid excursions and are thus valuable candidates for transplantation.
Collapse
Affiliation(s)
- Françoise Castex
- Biocommunication in Cardio-Metabolism (BC2M), University Montpellier, Montpellier, France
| | - Jeremy Leroy
- Biocommunication in Cardio-Metabolism (BC2M), University Montpellier, Montpellier, France
| | - Christophe Broca
- Laboratory of Cell Therapy for Diabetes, Institute for Regenerative Medicine & Biotherapy (IRMB), University Montpellier, INSERM, University Hospital Montpellier, Montpellier, France
| | - Karima Mezghenna
- Biocommunication in Cardio-Metabolism (BC2M), University Montpellier, Montpellier, France
| | - Flore Duranton
- Biocommunication in Cardio-Metabolism (BC2M), University Montpellier, Montpellier, France
- RD Néphrologie, Montpellier, France
| | - Vanessa Lavallard
- Department of Surgery, Cell Isolation and Transplantation Center, Geneva University Hospitals and University of Geneva, Genève, Switzerland
| | | | - René Gross
- Biocommunication in Cardio-Metabolism (BC2M), University Montpellier, Montpellier, France
| | - Anne Wojtusciszyn
- Laboratory of Cell Therapy for Diabetes, Institute for Regenerative Medicine & Biotherapy (IRMB), University Montpellier, INSERM, University Hospital Montpellier, Montpellier, France
| | - Anne-Dominique Lajoix
- Biocommunication in Cardio-Metabolism (BC2M), University Montpellier, Montpellier, France
| |
Collapse
|
10
|
Wang L, You ZH, Li YM, Zheng K, Huang YA. GCNCDA: A new method for predicting circRNA-disease associations based on Graph Convolutional Network Algorithm. PLoS Comput Biol 2020; 16:e1007568. [PMID: 32433655 PMCID: PMC7266350 DOI: 10.1371/journal.pcbi.1007568] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 06/02/2020] [Accepted: 03/23/2020] [Indexed: 01/22/2023] Open
Abstract
Numerous evidences indicate that Circular RNAs (circRNAs) are widely involved in the occurrence and development of diseases. Identifying the association between circRNAs and diseases plays a crucial role in exploring the pathogenesis of complex diseases and improving the diagnosis and treatment of diseases. However, due to the complex mechanisms between circRNAs and diseases, it is expensive and time-consuming to discover the new circRNA-disease associations by biological experiment. Therefore, there is increasingly urgent need for utilizing the computational methods to predict novel circRNA-disease associations. In this study, we propose a computational method called GCNCDA based on the deep learning Fast learning with Graph Convolutional Networks (FastGCN) algorithm to predict the potential disease-associated circRNAs. Specifically, the method first forms the unified descriptor by fusing disease semantic similarity information, disease and circRNA Gaussian Interaction Profile (GIP) kernel similarity information based on known circRNA-disease associations. The FastGCN algorithm is then used to objectively extract the high-level features contained in the fusion descriptor. Finally, the new circRNA-disease associations are accurately predicted by the Forest by Penalizing Attributes (Forest PA) classifier. The 5-fold cross-validation experiment of GCNCDA achieved 91.2% accuracy with 92.78% sensitivity at the AUC of 90.90% on circR2Disease benchmark dataset. In comparison with different classifier models, feature extraction models and other state-of-the-art methods, GCNCDA shows strong competitiveness. Furthermore, we conducted case study experiments on diseases including breast cancer, glioma and colorectal cancer. The results showed that 16, 15 and 17 of the top 20 candidate circRNAs with the highest prediction scores were respectively confirmed by relevant literature and databases. These results suggest that GCNCDA can effectively predict potential circRNA-disease associations and provide highly credible candidates for biological experiments. The recognition of circRNA-disease association is the key of disease diagnosis and treatment, and it is of great significance for exploring the pathogenesis of complex diseases. Computational methods can predict the potential disease-related circRNAs quickly and accurately. Based on the hypothesis that circRNA with similar function tends to associate with similar disease, GCNCDA model is proposed to effectively predict the potential association between circRNAs and diseases by combining FastGCN algorithm. The performance of the model was verified by cross-validation experiments, different feature extraction algorithm and classifier models comparison experiments. Furthermore, 16, 15 and 17 of the top 20 candidate circRNAs with the highest prediction scores in disease including breast cancer, glioma and colorectal cancer were respectively confirmed by relevant literature and databases. It is anticipated that GCNCDA model can give priority to the most promising circRNA-disease associations on a large scale to provide reliable candidates for further biological experiments.
Collapse
Affiliation(s)
- Lei Wang
- College of Information Science and Engineering, Zaozhuang University, Zaozhuang, China
- Xinjiang Technical Institutes of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, China
- * E-mail: (LW); (ZHY)
| | - Zhu-Hong You
- Xinjiang Technical Institutes of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, China
- * E-mail: (LW); (ZHY)
| | - Yang-Ming Li
- Department of Electrical Computer and Telecommunications Engineering Technology, Rochester Institute of Technology, Rochester, United States of America
| | - Kai Zheng
- School of Computer Science and Technology, China University of Mining and Technology, Xuzhou, China
| | - Yu-An Huang
- Department of Computing, Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
11
|
Liu Y, Deng J, Fan D. Ginsenoside Rk3 ameliorates high-fat-diet/streptozocin induced type 2 diabetes mellitus in mice via the AMPK/Akt signaling pathway. Food Funct 2020; 10:2538-2551. [PMID: 30993294 DOI: 10.1039/c9fo00095j] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Ginsenoside Rk3 (G-Rk3) is a main active ingredient of ginsenosides. Several recent studies demonstrated that ginsenosides have potential anti-type 2 diabetes mellitus (T2DM) properties. To evaluate the anti-T2DM effect of G-Rk3 and verify its potential mechanism, a high-fat-diet/streptozocin (HFD/STZ) induced model of T2DM in C57BL/6 mice and a high glucose induced insulin resistance model of HepG2 cells were applied in this research. Our analysis indicated that G-Rk3 reduced HFD/STZ induced hyperglycemia, and serum insulin and inflammation levels, and ameliorated glucose tolerance and insulin resistance, and prevented liver histological changes. Furthermore, it also significantly reduced lipid accumulation as shown by lower TG, LDL-C and TC serum concentrations and Oil Red O staining in liver tissues. The hypoglycemic effect of G-Rk3 seemed to be partially mediated via the inhibition of hepatic gluconeogenesis, which was supported by the activated p-Akt, p-FoxO1 and GLUT2 and inhibited FoxO1, PEPCK and G6pase protein expressions in the liver as well as increased glucose uptake in high glucose induced HepG2 cells. The gene expressions of hepatic gluconeogenesis were also down-regulated by G-Rk3 in HFD/STZ induced T2DM mice. In addition, G-Rk3 suppressed HFD/STZ induced lipid accumulation by regulating related gene and protein expressions such as p-ACC, FAS and SREBP-1, which are the downstream targets of AMPK. AMPK and Akt inhibitors significantly reversed G-Rk3 mediated hepatic gluconeogenesis and lipid accumulation. Thus, our study is the first to illustrate that G-Rk3 mediates hepatic gluconeogenesis and lipid accumulation via activating the AMPK/Akt signaling pathway in HFD/STZ induced T2DM mice.
Collapse
Affiliation(s)
- Yao Liu
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China.
| | | | | |
Collapse
|
12
|
Oleic acid increases the transcriptional activity of FoxO1 by promoting its nuclear translocation and β-catenin binding in pancreatic β-cells. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2753-2764. [PMID: 31255704 DOI: 10.1016/j.bbadis.2019.06.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/31/2019] [Accepted: 06/25/2019] [Indexed: 01/08/2023]
Abstract
In the setting of metabolic overload, chronic elevations of free fatty acids in blood and tissues are associated with pancreatic β-cell lipotoxicity and failure. Ultimately, obesity combined with insulin resistance increases the dysfunctional demand of β-cells and contributes to the development of type 2 diabetes. Forkhead box O1 (FoxO1) is a potent transcriptional regulator of pancreatic β-cell function and tolerance to lipid stress. The present study examined the effects of stearoyl-CoA desaturase 1 (SCD1)-metabolized precursors and products, notably oleic acid, on the compensatory capacity of β-cells and their relationship with regulation of the FoxO1 and Wnt pathways. The trioleate-induced compromise of insulin sensitivity blunted the compensatory response of pancreatic β-cells in primary rat islets. These events were associated with increases in the nuclear accumulation and transcriptional activity of FoxO1. Such effects were also observed in INS-1E cells that were subjected to oleate treatment. The overexpression of human SCD1 that was accompanied by endogenously generated oleic acid also led to an increase in the nuclear abundance of FoxO1. The mechanism of the oleate-mediated subcellular localization of FoxO1 was independent of the fatty acid receptor GPR40. Instead, the mechanism involved diversion of the active β-catenin pool from an interaction with transcription factor 7-like 2 toward FoxO1-mediated transcription in β-cells. Our findings identify a unique role for oleic acid in the compensatory response of pancreatic β-cells and emphasize the importance of FoxO1 in β-cell failure in obesity-induced insulin resistance.
Collapse
|
13
|
Abstract
Oxidative stress is one of the key mechanisms of sepsis related organ dysfunction including stress hyperglycemia. Silent mating type information regulation 2 homolog 1 (SIRT1) could regulate glucose metabolism through its deacetylase activity. In this study, we aimed to investigate the role of SIRT1/forkhead box protein 1 (FoxO1) pathway on lipopolysaccharide (LPS) induced INS-1 cells dysfunction from aspects of oxidative stress and apoptosis. After being treated with 1 mg/L LPS together with or without SIRT1 activator resveratrol (RSV) or SIRT1 inhibitor EX527, cell viability, ROS generation, malondialdehyde (MDA), superoxide, insulin secretion, and activity of superoxide dismutase (SOD) in INS-1 cells were measured by specific assays. Protein expression of SIRT1, FoxO1, toll-like receptor 4 (TLR4), and acetylated FoxO1 (ac-FoxO1) were detected by western blot analysis. Nuclear and cytoplasmic protein was extracted respectively to analyze SIRT1 and FoxO1 redistribution. Mitochondrial potentials and apoptosis were detected by flow cytometry or observed under fluorescence microscope. Results showed that LPS decreased cell viability and insulin secretion, increased ROS, MDA, and superoxide generation, whereas inhibited SOD activity and FoxO1 nuclear transportation. Activation of SIRT1 by RSV down-regulated TLR4 expression, SIRT1 and FoxO1 nuclear protein expression increased after RSV pretreatment. Additionally, LPS induced decreased mitochondrial membrane potentials and structural abnormalities, which could be partially reversed by RSV. SIRT1/FoxO1 may be one of potential targets which could resist against LPS-induced INS-1 cells from oxidative stress damage and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Xingxing Mo
- a Department of Emergency , Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Xiao Wang
- a Department of Emergency , Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Qinmin Ge
- a Department of Emergency , Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Fan Bian
- b Department of Nephrology , Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine , Shanghai , China
| |
Collapse
|
14
|
Maffei A, Lembo G, Carnevale D. PI3Kinases in Diabetes Mellitus and Its Related Complications. Int J Mol Sci 2018; 19:ijms19124098. [PMID: 30567315 PMCID: PMC6321267 DOI: 10.3390/ijms19124098] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/13/2018] [Accepted: 12/15/2018] [Indexed: 02/07/2023] Open
Abstract
Recent studies have shown that phosphoinositide 3-kinases (PI3Ks) have become the target of many pharmacological treatments, both in clinical trials and in clinical practice. PI3Ks play an important role in glucose regulation, and this suggests their possible involvement in the onset of diabetes mellitus. In this review, we gather our knowledge regarding the effects of PI3K isoforms on glucose regulation in several organs and on the most clinically-relevant complications of diabetes mellitus, such as cardiomyopathy, vasculopathy, nephropathy, and neurological disease. For instance, PI3K α has been proven to be protective against diabetes-induced heart failure, while PI3K γ inhibition is protective against the disease onset. In vessels, PI3K γ can generate oxidative stress, while PI3K β inhibition is anti-thrombotic. Finally, we describe the role of PI3Ks in Alzheimer’s disease and ADHD, discussing the relevance for diabetic patients. Given the high prevalence of diabetes mellitus, the multiple effects here described should be taken into account for the development and validation of drugs acting on PI3Ks.
Collapse
Affiliation(s)
- Angelo Maffei
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, 86077 Pozzilli, Italy.
| | - Giuseppe Lembo
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, 86077 Pozzilli, Italy.
- Department of Molecular Medicine, "Sapienza" University of Rome, 00161 Rome, Italy.
| | - Daniela Carnevale
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, 86077 Pozzilli, Italy.
- Department of Molecular Medicine, "Sapienza" University of Rome, 00161 Rome, Italy.
| |
Collapse
|
15
|
Zhang W, Wang L, Yu X, Jia A, Ming J, Ji Q. RFamide-related peptide-3 promotes alpha TC1 clone 6 cell survival likely via GPR147. Peptides 2018; 107:39-44. [PMID: 30081043 DOI: 10.1016/j.peptides.2018.07.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/08/2018] [Accepted: 07/31/2018] [Indexed: 12/19/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is closely related to islet alpha cell mass and viability. Several types of RFamide-related peptides (RFRPs) are involved in regulating proliferation and function of islet cells. However, current understanding of the role of RFamide-related peptide-3 (RFRP-3) in pancreatic alpha cells is limited. Therefore, we investigated the expression of the RFRP-3 receptor, G protein-coupled receptor 147 (GPR147), in mouse islets and alpha TC1 clone 6 cells, and evaluated the function of RFRP-3 on alpha cells. We show that GPR147 is expressed in mouse islets and alpha cell lines. In addition, RFRP-3 promotes survival of alpha cells under conditions of hyperglycemia and serum starvation. Mechanistic evidence demonstrates that RFRP-3 activated PI3K/AKT and ERK1/2 signaling cascades and treatment with an antagonist of GPR147, 1-adamantanecarbonyl-Arg-Phe-NH₂ (RF9) blocked this function. These findings indicate a novel effect of RFRP-3 in promoting alpha cell survival, likely via GPR147.
Collapse
Affiliation(s)
- Wencheng Zhang
- Department of Endocrinology, Xijing Hospital, Fourth Military Medical University, Xi'an, 169 Changle Road West, 710032, China
| | - Li Wang
- Department of Endocrinology, Xijing Hospital, Fourth Military Medical University, Xi'an, 169 Changle Road West, 710032, China
| | - Xinwen Yu
- Department of Endocrinology, Xijing Hospital, Fourth Military Medical University, Xi'an, 169 Changle Road West, 710032, China
| | - Aihua Jia
- Department of Endocrinology, Xijing Hospital, Fourth Military Medical University, Xi'an, 169 Changle Road West, 710032, China
| | - Jie Ming
- Department of Endocrinology, Xijing Hospital, Fourth Military Medical University, Xi'an, 169 Changle Road West, 710032, China
| | - Qiuhe Ji
- Department of Endocrinology, Xijing Hospital, Fourth Military Medical University, Xi'an, 169 Changle Road West, 710032, China.
| |
Collapse
|
16
|
Xu S, Zhou L, Ponnusamy M, Zhang L, Dong Y, Zhang Y, Wang Q, Liu J, Wang K. A comprehensive review of circRNA: from purification and identification to disease marker potential. PeerJ 2018; 6:e5503. [PMID: 30155370 PMCID: PMC6110255 DOI: 10.7717/peerj.5503] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/01/2018] [Indexed: 12/14/2022] Open
Abstract
Circular RNA (circRNA) is an endogenous noncoding RNA with a covalently closed cyclic structure. Based on their components, circRNAs are divided into exonic circRNAs, intronic circRNAs, and exon-intron circRNAs. CircRNAs have well-conserved sequences and often have high stability due to their resistance to exonucleases. Depending on their sequence, circRNAs are involved in different biological functions, including microRNA sponge activity, modulation of alternative splicing or transcription, interaction with RNA-binding proteins, and rolling translation, and are a derivative of pseudogenes. CircRNAs are involved in the development of a variety of pathological conditions, such as cardiovascular diseases, diabetes, neurological diseases, and cancer. Emerging evidence has shown that circRNAs are likely to be new potential clinical diagnostic markers or treatments for many diseases. Here we describe circRNA research methods and biological functions, and discuss the potential relationship between circRNAs and disease progression.
Collapse
Affiliation(s)
- Sheng Xu
- Center for Developmental Cardiology, Institute of Translational Medicine and School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - LuYu Zhou
- Center for Developmental Cardiology, Institute of Translational Medicine and School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Murugavel Ponnusamy
- Center for Developmental Cardiology, Institute of Translational Medicine and School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - LiXia Zhang
- Department of Inspection, The Medical Faculty of Qingdao University, Qingdao, Shandong, China
| | - YanHan Dong
- Center for Developmental Cardiology, Institute of Translational Medicine and School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - YanHui Zhang
- Center for Developmental Cardiology, Institute of Translational Medicine and School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Qi Wang
- Center for Developmental Cardiology, Institute of Translational Medicine and School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Jing Liu
- Center for Developmental Cardiology, Institute of Translational Medicine and School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Kun Wang
- Center for Developmental Cardiology, Institute of Translational Medicine and School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
17
|
Lawlor N, Youn A, Kursawe R, Ucar D, Stitzel ML. Alpha TC1 and Beta-TC-6 genomic profiling uncovers both shared and distinct transcriptional regulatory features with their primary islet counterparts. Sci Rep 2017; 7:11959. [PMID: 28931935 PMCID: PMC5607285 DOI: 10.1038/s41598-017-12335-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 09/06/2017] [Indexed: 01/09/2023] Open
Abstract
Alpha TC1 (αTC1) and Beta-TC-6 (βTC6) mouse islet cell lines are cellular models of islet (dys)function and type 2 diabetes (T2D). However, genomic characteristics of these cells, and their similarities to primary islet alpha and beta cells, are undefined. Here, we report the epigenomic (ATAC-seq) and transcriptomic (RNA-seq) landscapes of αTC1 and βTC6 cells. Each cell type exhibits hallmarks of its primary islet cell counterpart including cell-specific expression of beta (e.g., Pdx1) and alpha (e.g., Arx) cell transcription factors (TFs), and enrichment of binding motifs for these TFs in αTC1/βTC6 cis-regulatory elements. αTC1/βTC6 transcriptomes overlap significantly with the transcriptomes of primary mouse/human alpha and beta cells. Our data further indicate that ATAC-seq detects cell-specific regulatory elements for cell types comprising ≥ 20% of a mixed cell population. We identified αTC1/βTC6 cis-regulatory elements orthologous to those containing type 2 diabetes (T2D)-associated SNPs in human islets for 33 loci, suggesting these cells’ utility to dissect T2D molecular genetics in these regions. Together, these maps provide important insights into the conserved regulatory architecture between αTC1/βTC6 and primary islet cells that can be leveraged in functional (epi)genomic approaches to dissect the genetic and molecular factors controlling islet cell identity and function.
Collapse
Affiliation(s)
- Nathan Lawlor
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Ahrim Youn
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Romy Kursawe
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA. .,Institute for Systems Genomics, University of Connecticut, Farmington, CT, 06032, USA. .,Department of Genetics & Genome Sciences, University of Connecticut, Farmington, CT, 06032, USA.
| | - Michael L Stitzel
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA. .,Institute for Systems Genomics, University of Connecticut, Farmington, CT, 06032, USA. .,Department of Genetics & Genome Sciences, University of Connecticut, Farmington, CT, 06032, USA.
| |
Collapse
|