1
|
Bahardoust M, Hadaegh F, Mehrabi Y, Delpisheh A, Khalili D. Medication time of metformin and sulfonylureas and incidence of cardiovascular diseases and mortality in type 2 diabetes: a pooled cohort analysis. Sci Rep 2025; 15:8401. [PMID: 40069233 PMCID: PMC11897161 DOI: 10.1038/s41598-025-89721-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 02/07/2025] [Indexed: 03/15/2025] Open
Abstract
The effect of duration of medication with metformin and sulfonylurea (SUs) on cardiovascular diseases (CVD) and mortality events by duration of type 2 diabetes (DM) is unclear. This study aimed to investigate the effect of duration of treatment with metformin and SUs on CVD and mortality events based on DM duration in newly diagnosed DM patients. Data from three prospective cohorts of Tehran Lipid and Glucose Study (TLGS), Multi-Ethnic Study of Atherosclerosis (MESA), and Atherosclerosis Risk in Communities (ARIC) including 4108 newly diagnosed type 2 diabetes individuals (mean age, 59.4 ± 0.66 years) were pooled. Exposure was defined as the duration of metformin alone, SUs alone, and a combination of both since drug initiation. The Cox proportional hazards (CPH) model adjusted for confounders, including statin, aspirin, and anti-hypertensive, was used to estimate the hazard ratio (HR) (95% CI) for the outcomes. Cumulative exposure for metformin, SUs, aspirin, statin, and anti-hypertensive medication was calculated using the same method. The median follow-up was 20.33 ± 0.45 years. Cardiovascular events, all-cause mortality (ACM), and CVD mortality occurred in 767, 913, and 439 newly diagnosed DM patients, respectively. Metformin alone reduced the hazard of cardiovascular events, ACM, and CV-mortality by 7%, 4%, and 6%, respectively, for each year of use, respectively (p < 0.05); the corresponding values for SUs alone were 4%, 3%, and 4%, respectively (p < 0.05). The effect of metformin on reducing cardiovascular events, ACM, and CVD mortality continued until approximately 8, 10, and 5 years after the start of treatment, respectively, and then reached Plato. The effect of SUs on cardiovascular events, ACM, and CVD mortality continued to decline or reach Plato until approximately 6, 5, and 8 years after initiation of therapy and then was ineffective or reversed. The effect of the combination therapy on the reduction of cardiovascular events continued until 11 years after therapy initiation. Among newly diagnosed DM patients, metformin, with and without SUs, was associated with a reduced risk of cardiovascular events, ACM, and CVD mortality for up to about one decade. The combined effect of metformin + sulfonylurea was superior to the single effect of metformin or sulfonylurea alone. The combination therapy of Metformin and SUs can still be used with good safety, especially in the first years of diabetes diagnosis.
Collapse
Affiliation(s)
- Mansour Bahardoust
- Department of Epidemiology, School of Public Health & Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzad Hadaegh
- Prevention of Metabolic Disorders Research Center, Research Institute for Metabolic and Obesity Disorders, Research Institute for Endocrine Sciences, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yadollah Mehrabi
- Department of Epidemiology, School of Public Health & Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Delpisheh
- Department of Epidemiology, School of Public Health & Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Safety Promotion and Injury Prevention Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Davood Khalili
- Prevention of Metabolic Disorders Research Center, Research Institute for Metabolic and Obesity Disorders, Research Institute for Endocrine Sciences, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Lown Scholar in Cardiovascular Health, Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston,MA, US.
| |
Collapse
|
2
|
Zhang Y, Li M, Liu H, Fan Y, Liu HH. The application of procyanidins in diabetes and its complications: a review of preclinical studies. Front Pharmacol 2025; 16:1532246. [PMID: 39995417 PMCID: PMC11847907 DOI: 10.3389/fphar.2025.1532246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/14/2025] [Indexed: 02/26/2025] Open
Abstract
Diabetes mellitus (DM) and its various complications, including diabetic nephropathy, retinopathy, neuropathy, cardiovascular disease, and ulcers, pose significant challenges to global health. This review investigates the potential of procyanidins (PCs), a natural polyphenolic compound, in preventing and managing diabetes and its complications. PCs, recognized for their strong antioxidant, anti-inflammatory, and anti-hyperglycemic properties, play a crucial role in reducing oxidative stress and enhancing endothelial function, which are essential for managing diabetic complications. This review elucidates the molecular mechanisms by which PCs improve insulin sensitivity and endothelial health, thereby providing protection against the various complications of diabetes. The comprehensive analysis underscores the promising therapeutic role of PCs in diabetes care, indicating the need for further clinical studies to confirm and leverage their potential in comprehensive diabetes management strategies.
Collapse
Affiliation(s)
- Yongchuang Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengna Li
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong, China
| | - Haoyuan Liu
- Rehabilitation Department, Henan Provincial Hospital of Traditional Chinese Medicine, Zhengzhou, China
| | - Yongfu Fan
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Huan Huan Liu
- International institute for Traditional Chinese Medicine, Guanzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
3
|
Parlar MA, Mutlu H, Doğantekin B, Musaoğlu İS, Albayrakoğlu ND, Yavuz ML, Özbolat ZB, Kaplan M. The Association of Statin Therapy with Liver and Pancreatic Fat Fraction in Type 2 Diabetes Mellitus. Diagnostics (Basel) 2025; 15:426. [PMID: 40002577 PMCID: PMC11854770 DOI: 10.3390/diagnostics15040426] [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: 12/26/2024] [Revised: 02/02/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: It has been shown that the use of statins in patients with type 2 diabetes mellitus (T2DM) worsens hyperglycemia and hemoglobin A1c levels but may help in the preservation of pancreatic β-cell function. The potential role of a high pancreatic fat fraction (PFF) in this process has not yet been clarified. This study aimed to investigate whether the liver fat fraction (LFF) and PFF in T2DM patients is affected by statin therapy. Methods: This cross-sectional study involved a total of 140 T2DM patients, including both those who were receiving (n = 70) and those who were not receiving (n = 70) statin therapy. The mapping of the LFF and PFF utilizing the IDEAL-IQ sequence was conducted in magnetic resonance imaging. Results: In T2DM patients who used statins, the median PFF was higher compared to those who did not use statins (8.4 vs. 6.2%, p = 0.021), while the median LFF was found to be similar (8.4 vs. 8.9, p = 0.572). Variations in PFF were associated with the use of various statins (non-statin group: 6.2 vs. atovastatin: 8.7 vs. rosuvastatin: 3.2 vs. pitavastatin: 9.2, p = 0.004). The multivariable regression analysis indicated that insulin usage decreased log(LFF) by a factor of 0.16-fold (ꞵ ± SE = -0.16 ± 0.05, p = 0.010), and rosuvastatin usage reduced log(PFF) by 0.16-fold (ꞵ ± SE = -0.16 ± 0.07, p = 0.025), irrespective of other risk factors. Furthermore, the use of atorvastatin (ꞵ ± SE = 0.17 ± 0.06, p = 0.011) and pitavastatin (ꞵ ± SE = 0.19 ± 0.07, p = 0.008) were independently associated with an increase in log(PFF). Conclusions: In patients with T2DM, statin use did not show a significant effect on the liver fat fraction, but it caused differences in the pancreatic fat fraction. The observation of a lower pancreatic fat fraction in patients taking a rosuvastatin and atorvastatin dose of 40 mg/day suggests that different types and doses of statins may have varying effects on pancreatic fat accumulation.
Collapse
Affiliation(s)
- Mehmet Akif Parlar
- Department of Internal Medicine, Sultan 2. Abdülhamid Han Training and Research Hospital, University of Health Sciences, Selimiye Neighborhood, Tıbbiye Street, 34668 Istanbul, Turkey; (H.M.); (B.D.); (İ.S.M.); (N.D.A.); (M.K.)
| | - Hakan Mutlu
- Department of Internal Medicine, Sultan 2. Abdülhamid Han Training and Research Hospital, University of Health Sciences, Selimiye Neighborhood, Tıbbiye Street, 34668 Istanbul, Turkey; (H.M.); (B.D.); (İ.S.M.); (N.D.A.); (M.K.)
| | - Betül Doğantekin
- Department of Internal Medicine, Sultan 2. Abdülhamid Han Training and Research Hospital, University of Health Sciences, Selimiye Neighborhood, Tıbbiye Street, 34668 Istanbul, Turkey; (H.M.); (B.D.); (İ.S.M.); (N.D.A.); (M.K.)
| | - İsmail Serhat Musaoğlu
- Department of Internal Medicine, Sultan 2. Abdülhamid Han Training and Research Hospital, University of Health Sciences, Selimiye Neighborhood, Tıbbiye Street, 34668 Istanbul, Turkey; (H.M.); (B.D.); (İ.S.M.); (N.D.A.); (M.K.)
| | - Nisa Demirboşnak Albayrakoğlu
- Department of Internal Medicine, Sultan 2. Abdülhamid Han Training and Research Hospital, University of Health Sciences, Selimiye Neighborhood, Tıbbiye Street, 34668 Istanbul, Turkey; (H.M.); (B.D.); (İ.S.M.); (N.D.A.); (M.K.)
| | - Mustafa Lütfi Yavuz
- Department of Cardiology, Istanbul University Faculty of Medicine, 34093 Istanbul, Turkey;
| | - Zehra Buşra Özbolat
- Deparment of Chest Diseases, Çerkezköy State Hospital, Tekirdağ Provincial Health Directorate, 59100 Tekirdağ, Turkey;
| | - Mustafa Kaplan
- Department of Internal Medicine, Sultan 2. Abdülhamid Han Training and Research Hospital, University of Health Sciences, Selimiye Neighborhood, Tıbbiye Street, 34668 Istanbul, Turkey; (H.M.); (B.D.); (İ.S.M.); (N.D.A.); (M.K.)
| |
Collapse
|
4
|
Zhou J, Shi Y, Zhao L, Wang R, Luo L, Yin Z. γ-Glutamylcysteine restores glucolipotoxicity-induced islet β-cell apoptosis and dysfunction via inhibiting endoplasmic reticulum stress. Toxicol Appl Pharmacol 2025; 495:117206. [PMID: 39701215 DOI: 10.1016/j.taap.2024.117206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/11/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024]
Abstract
PURPOSE The impaired function of islet β-cell is associated with the pathogenesis of type 2 diabetes mellitus (T2DM). γ-glutamylcysteine (γ-GC), an immediate precursor of glutathione (GSH), has antioxidant and neuroprotective functions. Its level has been reported to be down-regulated in hyperglycemia. However, whether γ-GC has a protective effect on islet β-cell dysfunction remains elusive. Recently, we explore the molecular mechanism by which γ-GC protects islet β-cell from glucolipotoxicity-induced dysfunction. METHODS In vivo mice models and in vitro cell models were established to examine the therapeutic effects and molecular mechanisms of γ-GC. RESULTS db mice develop impaired glucose-stimulated insulin secretion (GSIS) due to reduced islet number and damaged islet microstructure. Serious oxidative damage, apoptosis and lipid accumulation are also observed in β-cell stimulated by glucolipotoxicity. Mechanistic studies suggest that glucolipotoxicity inhibits PDX-1 nuclear translocation by inducing endoplasmic reticulum (ER) stress, which leads to impaired insulin (INS) secretion in β-cell. Nevertheless, γ-GC as an inhibitor of ER stress can alleviate the damage of islet microstructure in db mice. Importantly, γ-GC promotes INS gene expression and GSIS through driving nuclear translocation of PDX-1, thereby enhancing intracellular INS content. Moreover, treatment with γ-GC can also mitigate oxidative damage, apoptosis and lipid accumulation of β-cell, resulting in ameliorating islet β-cell dysfunction induced by glucolipotoxicity. CONCLUSION Our results support the use of γ-GC as an inhibitor of ER stress for prevention and treatment of T2DM in the future.
Collapse
Affiliation(s)
- Jinyi Zhou
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Yingying Shi
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Lishuang Zhao
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Rong Wang
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Lan Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.
| | - Zhimin Yin
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China.
| |
Collapse
|
5
|
Huang C, Chen X, Ouyang Z, Meng L, Liu J, Pang Q, Fan R. Bisphenol a accelerates the glucolipotoxicity-induced dysfunction of rat insulinoma cell lines: An implication for a potential risk of environmental bisphenol a exposure for individuals susceptible to type 2 diabetes. Toxicol In Vitro 2024; 99:105866. [PMID: 38844119 DOI: 10.1016/j.tiv.2024.105866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/21/2024] [Accepted: 06/03/2024] [Indexed: 06/11/2024]
Abstract
Epidemiological studies have suggested a correlation between bisphenol A (BPA) and type 2 diabetes (T2DM). The effects of BPA on β-cell dysfunction may reveal the risks from an in vitro perspective. We used the rat insulinoma (INS-1) cell lines (a type of β-cells) to set up normal or damaged models (DM), which were exposed to various concentrations of BPA (0.001, 0.01, 0.1, 1, 10 and 100 μM). An increase in reactive oxygen species (ROS) and apoptosis, and a decrease in cell viability were observed in INS-1 cells exposed to high doses of BPA for 48 h. Interestingly, exposure to lower doses of BPA for 24 h resulted in increased ROS levels and apoptosis rates in INS-1 in the DM group, along with decreased cell viability, suggesting that BPA exerts toxicity to INS-1 cells, particularly to the DM group. Insulin levels and Glut2 expression, glucose consumption, intracellular Ca2+ and insulin secretion were increased in INS-1 cells after 48 h exposure to high dose of BPA. Stronger effects were observed in the DM group, even those exposed to low doses of BPA for 24 h. Moreover, BPA inhibited high glucose-stimulated insulin secretion in these cells. Our research suggests that low doses of BPA exacerbate the dysfunction caused by glucolipotoxicity, implying environmental BPA exposure poses a risk for individuals with prediabetes or T2DM.
Collapse
Affiliation(s)
- Chengmeng Huang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Xiaolin Chen
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Zedong Ouyang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Lingxue Meng
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Jian Liu
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Qihua Pang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China.
| | - Ruifang Fan
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
6
|
Perrier J, Nawrot M, Madec AM, Chikh K, Chauvin MA, Damblon C, Sabatier J, Thivolet CH, Rieusset J, Rautureau GJP, Panthu B. Human Pancreatic Islets React to Glucolipotoxicity by Secreting Pyruvate and Citrate. Nutrients 2023; 15:4791. [PMID: 38004183 PMCID: PMC10674605 DOI: 10.3390/nu15224791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Progressive decline in pancreatic beta-cell function is central to the pathogenesis of type 2 diabetes (T2D). Here, we explore the relationship between the beta cell and its nutritional environment, asking how an excess of energy substrate leads to altered energy production and subsequent insulin secretion. Alterations in intracellular metabolic homeostasis are key markers of islets with T2D, but changes in cellular metabolite exchanges with their environment remain unknown. We answered this question using nuclear magnetic resonance-based quantitative metabolomics and evaluated the consumption or secretion of 31 extracellular metabolites from healthy and T2D human islets. Islets were also cultured under high levels of glucose and/or palmitate to induce gluco-, lipo-, and glucolipotoxicity. Biochemical analyses revealed drastic alterations in the pyruvate and citrate pathways, which appear to be associated with mitochondrial oxoglutarate dehydrogenase (OGDH) downregulation. We repeated these manipulations on the rat insulinoma-derived beta-pancreatic cell line (INS-1E). Our results highlight an OGDH downregulation with a clear effect on the pyruvate and citrate pathways. However, citrate is directed to lipogenesis in the INS-1E cells instead of being secreted as in human islets. Our results demonstrate the ability of metabolomic approaches performed on culture media to easily discriminate T2D from healthy and functional islets.
Collapse
Affiliation(s)
- Johan Perrier
- Laboratoire CarMeN, UMR INSERM U1060/INRAE U1397, University of Lyon, Université Claude Bernard Lyon 1, 69310 Pierre-Bénite, France
| | - Margaux Nawrot
- Laboratoire CarMeN, UMR INSERM U1060/INRAE U1397, University of Lyon, Université Claude Bernard Lyon 1, 69310 Pierre-Bénite, France
| | - Anne-Marie Madec
- Laboratoire CarMeN, UMR INSERM U1060/INRAE U1397, University of Lyon, Université Claude Bernard Lyon 1, 69310 Pierre-Bénite, France
| | - Karim Chikh
- Laboratoire CarMeN, UMR INSERM U1060/INRAE U1397, University of Lyon, Université Claude Bernard Lyon 1, 69310 Pierre-Bénite, France
- Department of Endocrinology and Diabetes, Hospices Civils de Lyon, Hopital Lyon Sud, 69310 Pierre-Bénite, France
| | - Marie-Agnès Chauvin
- Laboratoire CarMeN, UMR INSERM U1060/INRAE U1397, University of Lyon, Université Claude Bernard Lyon 1, 69310 Pierre-Bénite, France
| | - Christian Damblon
- Unité de Recherche MolSys, Faculté des Sciences, Université de Liège, 99131 Liège, Belgium
| | - Julia Sabatier
- Laboratory of Cell Therapy for Diabetes (LTCD), PRIMS Facility, Institute for Regenerative Medicine and Biotherapy (IRMB), University Hospital of Montpellier, 34295 Montpellier, France
| | - Charles H. Thivolet
- Laboratoire CarMeN, UMR INSERM U1060/INRAE U1397, University of Lyon, Université Claude Bernard Lyon 1, 69310 Pierre-Bénite, France
- Department of Endocrinology and Diabetes, Hospices Civils de Lyon, Hopital Lyon Sud, 69310 Pierre-Bénite, France
| | - Jennifer Rieusset
- Laboratoire CarMeN, UMR INSERM U1060/INRAE U1397, University of Lyon, Université Claude Bernard Lyon 1, 69310 Pierre-Bénite, France
| | - Gilles J. P. Rautureau
- Centre de Résonance Magnétique Nucléaire à Très Hauts Champs, UMR 5082 CNRS, ENS Lyon, UCBL, Université de Lyon, 69100 Villeurbanne, France
| | - Baptiste Panthu
- Laboratoire CarMeN, UMR INSERM U1060/INRAE U1397, University of Lyon, Université Claude Bernard Lyon 1, 69310 Pierre-Bénite, France
| |
Collapse
|
7
|
St. Clair JR, Westacott MJ, Miranda J, Farnsworth NL, Kravets V, Schleicher WE, Dwulet JM, Levitt CH, Heintz A, Ludin NWF, Benninger RKP. Restoring connexin-36 function in diabetogenic environments precludes mouse and human islet dysfunction. J Physiol 2023; 601:4053-4072. [PMID: 37578890 PMCID: PMC10508056 DOI: 10.1113/jp282114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 07/27/2023] [Indexed: 08/16/2023] Open
Abstract
The secretion of insulin from β-cells in the islet of Langerhans is governed by a series of metabolic and electrical events, which can fail during the progression of type 2 diabetes (T2D). β-cells are electrically coupled via connexin-36 (Cx36) gap junction channels, which coordinates the pulsatile dynamics of [Ca2+ ] and insulin release across the islet. Factors such as pro-inflammatory cytokines and free fatty acids disrupt gap junction coupling under in vitro conditions. Here we test whether gap junction coupling and coordinated [Ca2+ ] dynamics are disrupted in T2D, and whether recovery of gap junction coupling can recover islet function. We examine islets from donors with T2D, from db/db mice, and islets treated with pro-inflammatory cytokines (TNF-α, IL-1β, IFN-ɣ) or free fatty acids (palmitate). We modulate gap junction coupling using Cx36 over-expression or pharmacological activation via modafinil. We also develop a peptide mimetic (S293) of the c-terminal regulatory site of Cx36 designed to compete against its phosphorylation. Cx36 gap junction permeability and [Ca2+ ] dynamics were disrupted in islets from both human donors with T2D and db/db mice, and in islets treated with pro-inflammatory cytokines or palmitate. Cx36 over-expression, modafinil treatment and S293 peptide all enhanced Cx36 gap junction coupling and protected against declines in coordinated [Ca2+ ] dynamics. Cx36 over-expression and S293 peptide also reduced apoptosis induced by pro-inflammatory cytokines. Critically, S293 peptide rescued gap junction coupling and [Ca2+ ] dynamics in islets from both db/db mice and a sub-set of T2D donors. Thus, recovering or enhancing Cx36 gap junction coupling can improve islet function in diabetes. KEY POINTS: Connexin-36 (Cx36) gap junction permeability and associated coordination of [Ca2+ ] dynamics is diminished in human type 2 diabetes (T2D) and mouse models of T2D. Enhancing Cx36 gap junction permeability protects against disruptions to the coordination of [Ca2+ ] dynamics. A novel peptide mimetic of the Cx36 c-terminal regulatory region protects against declines in Cx36 gap junction permeability. Pharmacological elevation in Cx36 or Cx36 peptide mimetic recovers [Ca2+ ] dynamics and glucose-stimulated insulin secretion in human T2D and mouse models of T2D.
Collapse
Affiliation(s)
- Joshua R St. Clair
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Matthew J Westacott
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Jose Miranda
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Nikki L Farnsworth
- Barbara Davis Center for Diabetes, University of Colorado
Denver | Anschutz Medical Campus, Aurora, CO
| | - Vira Kravets
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Wolfgang E Schleicher
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - JaeAnn M Dwulet
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Claire H Levitt
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Audrey Heintz
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Nurin WF Ludin
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Richard KP Benninger
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
- Barbara Davis Center for Diabetes, University of Colorado
Denver | Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
8
|
Tariq M, de Souza AH, Bensellam M, Chae H, Jaffredo M, Close AF, Deglasse JP, Santos LRB, Buemi A, Mourad NI, Wojtusciszyn A, Raoux M, Gilon P, Broca C, Jonas JC. Prolonged culture of human pancreatic islets under glucotoxic conditions changes their acute beta cell calcium and insulin secretion glucose response curves from sigmoid to bell-shaped. Diabetologia 2023; 66:709-723. [PMID: 36459178 DOI: 10.1007/s00125-022-05842-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/18/2022] [Indexed: 12/04/2022]
Abstract
AIMS/HYPOTHESIS The rapid remission of type 2 diabetes by a diet very low in energy correlates with a marked improvement in glucose-stimulated insulin secretion (GSIS), emphasising the role of beta cell dysfunction in the early stages of the disease. In search of novel mechanisms of beta cell dysfunction after long-term exposure to mild to severe glucotoxic conditions, we extensively characterised the alterations in insulin secretion and upstream coupling events in human islets cultured for 1-3 weeks at ~5, 8, 10 or 20 mmol/l glucose and subsequently stimulated by an acute stepwise increase in glucose concentration. METHODS Human islets from 49 non-diabetic donors (ND-islets) and six type 2 diabetic donors (T2D-islets) were obtained from five isolation centres. After shipment, the islets were precultured for 3-7 days in RPMI medium containing ~5 mmol/l glucose and 10% (vol/vol) heat-inactivated FBS with selective islet picking at each medium renewal. Islets were then cultured for 1-3 weeks in RPMI containing ~5, 8, 10 or 20 mmol/l glucose before measurement of insulin secretion during culture, islet insulin and DNA content, beta cell apoptosis and cytosolic and mitochondrial glutathione redox state, and assessment of dynamic insulin secretion and upstream coupling events during acute stepwise stimulation with glucose [NAD(P)H autofluorescence, ATP/(ATP+ADP) ratio, electrical activity, cytosolic Ca2+ concentration ([Ca2+]c)]. RESULTS Culture of ND-islets for 1-3 weeks at 8, 10 or 20 vs 5 mmol/l glucose did not significantly increase beta cell apoptosis or oxidative stress but decreased insulin content in a concentration-dependent manner and increased beta cell sensitivity to subsequent acute stimulation with glucose. Islet glucose responsiveness was higher after culture at 8 or 10 vs 5 mmol/l glucose and markedly reduced after culture at 20 vs 5 mmol/l glucose. In addition, the [Ca2+]c and insulin secretion responses to acute stepwise stimulation with glucose were no longer sigmoid but bell-shaped, with maximal stimulation at 5 or 10 mmol/l glucose and rapid sustained inhibition above that concentration. Such paradoxical inhibition was, however, no longer observed when islets were acutely depolarised by 30 mmol/l extracellular K+. The glucotoxic alterations of beta cell function were fully reversible after culture at 5 mmol/l glucose and were mimicked by pharmacological activation of glucokinase during culture at 5 mmol/l glucose. Similar results to those seen in ND-islets were obtained in T2D-islets, except that their rate of insulin secretion during culture at 8 and 20 mmol/l glucose was lower, their cytosolic glutathione oxidation increased after culture at 8 and 20 mmol/l glucose, and the alterations in GSIS and upstream coupling events were greater after culture at 8 mmol/l glucose. CONCLUSIONS/INTERPRETATION Prolonged culture of human islets under moderate to severe glucotoxic conditions markedly increased their glucose sensitivity and revealed a bell-shaped acute glucose response curve for changes in [Ca2+]c and insulin secretion, with maximal stimulation at 5 or 10 mmol/l glucose and rapid inhibition above that concentration. This novel glucotoxic alteration may contribute to beta cell dysfunction in type 2 diabetes independently from a detectable increase in beta cell apoptosis.
Collapse
Affiliation(s)
- Mohammad Tariq
- Secteur des sciences de la santé, Institut de recherche expérimentale et clinique, Pôle d'endocrinologie, diabète et nutrition, Université catholique de Louvain, Brussels, Belgium
- Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Arnaldo H de Souza
- Secteur des sciences de la santé, Institut de recherche expérimentale et clinique, Pôle d'endocrinologie, diabète et nutrition, Université catholique de Louvain, Brussels, Belgium
| | - Mohammed Bensellam
- Secteur des sciences de la santé, Institut de recherche expérimentale et clinique, Pôle d'endocrinologie, diabète et nutrition, Université catholique de Louvain, Brussels, Belgium
| | - Heeyoung Chae
- Secteur des sciences de la santé, Institut de recherche expérimentale et clinique, Pôle d'endocrinologie, diabète et nutrition, Université catholique de Louvain, Brussels, Belgium
| | - Manon Jaffredo
- CNRS, Institute of Chemistry and Biology of Membranes and Nano-objects, UMR 5248, University of Bordeaux, Pessac, France
| | - Anne-Françoise Close
- Secteur des sciences de la santé, Institut de recherche expérimentale et clinique, Pôle d'endocrinologie, diabète et nutrition, Université catholique de Louvain, Brussels, Belgium
| | - Jean-Philippe Deglasse
- Secteur des sciences de la santé, Institut de recherche expérimentale et clinique, Pôle d'endocrinologie, diabète et nutrition, Université catholique de Louvain, Brussels, Belgium
| | - Laila R B Santos
- Secteur des sciences de la santé, Institut de recherche expérimentale et clinique, Pôle d'endocrinologie, diabète et nutrition, Université catholique de Louvain, Brussels, Belgium
- Bio Base Europe Pilot Plant (BBEPP), Ghent, Belgium
| | - Antoine Buemi
- Secteur des sciences de la santé, Institut de recherche expérimentale et clinique, Pôle de chirurgie expérimentale, Université catholique de Louvain, Brussels, Belgium
| | - Nizar I Mourad
- Secteur des sciences de la santé, Institut de recherche expérimentale et clinique, Pôle de chirurgie expérimentale, Université catholique de Louvain, Brussels, Belgium
| | - Anne Wojtusciszyn
- Laboratoire de Thérapie Cellulaire du Diabète, Institut de Médecine Régénérative et Biothérapies, Hôpital St Eloi, CHU Montpellier, Montpellier, France
- Service d'Endocrinologie, Diabétologie et Métabolisme, Centre Hospitalier Universitaire Vaudois and Université de Lausanne, Lausanne, Switzerland
| | - Matthieu Raoux
- CNRS, Institute of Chemistry and Biology of Membranes and Nano-objects, UMR 5248, University of Bordeaux, Pessac, France
| | - Patrick Gilon
- Secteur des sciences de la santé, Institut de recherche expérimentale et clinique, Pôle d'endocrinologie, diabète et nutrition, Université catholique de Louvain, Brussels, Belgium
| | - Christophe Broca
- Laboratoire de Thérapie Cellulaire du Diabète, Institut de Médecine Régénérative et Biothérapies, Hôpital St Eloi, CHU Montpellier, Montpellier, France
| | - Jean-Christophe Jonas
- Secteur des sciences de la santé, Institut de recherche expérimentale et clinique, Pôle d'endocrinologie, diabète et nutrition, Université catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
9
|
Zhang R, Zhao Q, Li R. Predictors of acarbose therapeutic efficacy in newly diagnosed type 2 diabetes mellitus patients in China. BMC Pharmacol Toxicol 2022; 23:79. [PMID: 36258236 PMCID: PMC9580108 DOI: 10.1186/s40360-022-00621-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 10/10/2022] [Indexed: 11/10/2022] Open
Abstract
Background Acarbose is one of the optimal drugs for patients with the first diagnosis of type 2 diabetes mellitus (T2DM). But what kind of emerging patients has the best therapeutic response to acarbose therapy has never been reported. To this end, we investigated predictors of acarbose therapeutic efficacy in newly diagnosed T2DM patients in China. Methods A total of 346 T2DM patients received acarbose monotherapy for 48 weeks as part of participating in the Study of Acarbose in Newly Diagnosed Patients with T2DM in China (MARCH study) from November 2008 to June 2011. Change in glycated hemoglobin (ΔHbA1c) served as a dependent variable while different baseline variables including sex, age, disease duration, weight, body mass index (BMI), systolic blood pressure (SBP), diastolic blood pressure (DBP), HbA1c, fasting plasma glucose (FPG), 2-h postprandial blood glucose (2 h PG), fasting insulin (FINS), 2-h postprandial insulin (2 h INS), early insulin secretion index (IGI), homeostasis model assessment of insulin resistance index (HOMA-IR), homeostasis model assessment of beta cell function (HOMA-B), area under the curve (AUC) of glucagon, insulin and GLP-1 were assessed as independent predictors. Step-wise multiple linear regression was employed for statistical analysis. Results The results suggested that independent predictors of ΔHbA1c at 12 weeks included baseline body weight (β = − 0.012, P = 0.006), DBP (β = 0.010, P = 0.047), FPG (β = 0.111, P = 0.005) and 2 h PG (β = 0.042, P = 0.043). Independent predictors of ΔHbA1c at 24 weeks included disease duration (β = 0.040, P = 0.019) and FPG (β = 0.117, P = 0.001). Finally, independent predictor of ΔHbA1c at 48 weeks was disease duration (β = 0.038, P = 0.046). Conclusions Acarbose may be more effective in newly diagnosed T2DM patients with low FPG, low 2 h PG and obesity. The earlier T2DM is diagnosed and continuously treated with acarbose, the better the response to therapy.
Collapse
Affiliation(s)
- Rong Zhang
- Department of Geriatrics, Xijing Hospital of Air Force Medical University, No.127 West Changle Road, Xi'an, 710032, China
| | - Quanxi Zhao
- Department of Pharmacy, Second People's Hospital of Shaanxi Province, No.3 Shangqin Road, Xi'an, 710068, China
| | - Rong Li
- Department of Geriatrics, Xijing Hospital of Air Force Medical University, No.127 West Changle Road, Xi'an, 710032, China.
| |
Collapse
|
10
|
Jain C, Bilekova S, Lickert H. Targeting pancreatic β cells for diabetes treatment. Nat Metab 2022; 4:1097-1108. [PMID: 36131204 DOI: 10.1038/s42255-022-00618-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 07/13/2022] [Indexed: 11/09/2022]
Abstract
Insulin is a life-saving drug for patients with type 1 diabetes; however, even today, no pharmacotherapy can prevent the loss or dysfunction of pancreatic insulin-producing β cells to stop or reverse disease progression. Thus, pancreatic β cells have been a main focus for cell-replacement and regenerative therapies as a curative treatment for diabetes. In this Review, we highlight recent advances toward the development of diabetes therapies that target β cells to enhance proliferation, redifferentiation and protection from cell death and/or enable selective killing of senescent β cells. We describe currently available therapies and their mode of action, as well as insufficiencies of glucagon-like peptide 1 (GLP-1) and insulin therapies. We discuss and summarize data collected over the last decades that support the notion that pharmacological targeting of β cell insulin signalling might protect and/or regenerate β cells as an improved treatment of patients with diabetes.
Collapse
Affiliation(s)
- Chirag Jain
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Immunology Discovery, Genentech Inc., South San Francisco, CA, USA
| | - Sara Bilekova
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Chair of β-Cell Biology, Technische Universität München, School of Medicine, Klinikum Rechts der Isar, München, Germany.
| |
Collapse
|
11
|
McCarty MF, DiNicolantonio JJ. Maintaining Effective Beta Cell Function in the Face of Metabolic Syndrome-Associated Glucolipotoxicity-Nutraceutical Options. Healthcare (Basel) 2021; 10:3. [PMID: 35052168 PMCID: PMC8775473 DOI: 10.3390/healthcare10010003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 11/16/2022] Open
Abstract
In people with metabolic syndrome, episodic exposure of pancreatic beta cells to elevated levels of both glucose and free fatty acids (FFAs)-or glucolipotoxicity-can induce a loss of glucose-stimulated insulin secretion (GSIS). This in turn can lead to a chronic state of glucolipotoxicity and a sustained loss of GSIS, ushering in type 2 diabetes. Loss of GSIS reflects a decline in beta cell glucokinase (GK) expression associated with decreased nuclear levels of the pancreatic and duodenal homeobox 1 (PDX1) factor that drives its transcription, along with that of Glut2 and insulin. Glucolipotoxicity-induced production of reactive oxygen species (ROS), stemming from both mitochondria and the NOX2 isoform of NADPH oxidase, drives an increase in c-Jun N-terminal kinase (JNK) activity that promotes nuclear export of PDX1, and impairs autocrine insulin signaling; the latter effect decreases PDX1 expression at the transcriptional level and up-regulates beta cell apoptosis. Conversely, the incretin hormone glucagon-like peptide-1 (GLP-1) promotes nuclear import of PDX1 via cAMP signaling. Nutraceuticals that quell an increase in beta cell ROS production, that amplify or mimic autocrine insulin signaling, or that boost GLP-1 production, should help to maintain GSIS and suppress beta cell apoptosis in the face of glucolipotoxicity, postponing or preventing onset of type 2 diabetes. Nutraceuticals with potential in this regard include the following: phycocyanobilin-an inhibitor of NOX2; agents promoting mitophagy and mitochondrial biogenesis, such as ferulic acid, lipoic acid, melatonin, berberine, and astaxanthin; myo-inositol and high-dose biotin, which promote phosphatidylinositol 3-kinase (PI3K)/Akt activation; and prebiotics/probiotics capable of boosting GLP-1 secretion. Complex supplements or functional foods providing a selection of these agents might be useful for diabetes prevention.
Collapse
Affiliation(s)
| | - James J. DiNicolantonio
- Department of Preventive Cardiology, Saint Luke’s Mid America Heart Institute, Kansas City, MO 64111, USA
| |
Collapse
|
12
|
De Paoli M, Zakharia A, Werstuck GH. The Role of Estrogen in Insulin Resistance: A Review of Clinical and Preclinical Data. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1490-1498. [PMID: 34102108 DOI: 10.1016/j.ajpath.2021.05.011] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/10/2021] [Accepted: 05/18/2021] [Indexed: 12/26/2022]
Abstract
Insulin resistance results when peripheral tissues, including adipose, skeletal muscle, and liver, do not respond appropriately to insulin, causing the ineffective uptake of glucose. This represents a risk factor for the development of type 2 diabetes mellitus. Along with abdominal obesity, hypertension, high levels of triglycerides, and low levels of high-density lipoproteins, insulin resistance is a component of a condition known as the metabolic syndrome, which significantly increases the risk of developing cardiometabolic disorders. Accumulating evidence shows that biological sex has a major influence in the development of cardiometabolic disturbances, with females being more protected than males. This protection appears to be driven by female sex hormones (estrogens), as it tends to disappear with the onset of menopause but can be re-established with hormone replacement therapy. This review evaluates current knowledge on the protective role of estrogens in the relevant pathways associated with insulin resistance. The importance of increasing our understanding of sex as a biological variable in cardiometabolic research to promote the development of more effective preventative strategies is emphasized.
Collapse
Affiliation(s)
- Monica De Paoli
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Alexander Zakharia
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Geoff H Werstuck
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
13
|
Zhang L, Huang YJ, Sun JP, Zhang TY, Liu TL, Ke B, Shi XF, Li H, Zhang GP, Ye ZY, Hu J, Qin J. Protective effects of calorie restriction on insulin resistance and islet function in STZ-induced type 2 diabetes rats. Nutr Metab (Lond) 2021; 18:48. [PMID: 33952301 PMCID: PMC8097947 DOI: 10.1186/s12986-021-00575-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 04/16/2021] [Indexed: 12/26/2022] Open
Abstract
Background Caloric restriction (CR) has become increasingly attractive in the treatment of type 2 diabetes mellitus (T2DM) because of the increasingly common high-calorie diet and sedentary lifestyle. This study aimed to evaluate the role of CR in T2DM treatment and further explore its potential molecular mechanisms. Methods Sixty male Sprague–Dawley rats were used in this study. The diabetes model was induced by 8 weeks of high-fat diet (HFD) followed by a single dose of streptozotocin injection (30 mg/kg). Subsequently, the diabetic rats were fed HFD at 28 g/day (diabetic control) or 20 g/day (30% CR regimen) for 20 weeks. Meanwhile, normal rats fed a free standard chow diet served as the vehicle control. Body mass, plasma glucose levels, and lipid profiles were monitored. After diabetes-related functional tests were performed, the rats were sacrificed at 10 and 20 weeks, and glucose uptake in fresh muscle was determined. In addition, western blotting and immunofluorescence were used to detect alterations in AKT/AS160/GLUT4 signaling. Results We found that 30% CR significantly attenuated hyperglycemia and dyslipidemia, leading to alleviation of glucolipotoxicity and thus protection of islet function. Insulin resistance was also markedly ameliorated, as indicated by notably improved insulin tolerance and homeostatic model assessment for insulin resistance (HOMA-IR). However, the improvement in glucose uptake in skeletal muscle was not significant. The upregulation of AKT/AS160/GLUT4 signaling in muscle induced by 30% CR also attenuated gradually over time. Interestingly, the consecutive decrease in AKT/AS160/GLUT4 signaling in white adipose tissue was significantly reversed by 30% CR. Conclusion CR (30%) could protect islet function from hyperglycemia and dyslipidemia, and improve insulin resistance. The mechanism by which these effects occurred is likely related to the upregulation of AKT/AS160/GLUT4 signaling. Supplementary Information The online version contains supplementary material available at 10.1186/s12986-021-00575-y.
Collapse
Affiliation(s)
- Li Zhang
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.,Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 518100, China
| | - Ying-Juan Huang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 518100, China
| | - Jia-Pan Sun
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 518100, China
| | - Ting-Ying Zhang
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Tao-Li Liu
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Bin Ke
- Department of VIP Ward, Sun Yat-Sen University Cancer Center, Guangzhou, 510080, China
| | - Xian-Fang Shi
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Hui Li
- Department of Obese and Metabolic Disease, Guangzhou Panyu Hospital of Chinese Medicine, Guangzhou, 511400, China
| | - Geng-Peng Zhang
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Zhi-Yu Ye
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Jianguo Hu
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Jian Qin
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|
14
|
Elsayed AK, Vimalraj S, Nandakumar M, Abdelalim EM. Insulin resistance in diabetes: The promise of using induced pluripotent stem cell technology. World J Stem Cells 2021; 13:221-235. [PMID: 33815671 PMCID: PMC8006014 DOI: 10.4252/wjsc.v13.i3.221] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/07/2021] [Accepted: 03/11/2021] [Indexed: 02/06/2023] Open
Abstract
Insulin resistance (IR) is associated with several metabolic disorders, including type 2 diabetes (T2D). The development of IR in insulin target tissues involves genetic and acquired factors. Persons at genetic risk for T2D tend to develop IR several years before glucose intolerance. Several rodent models for both IR and T2D are being used to study the disease pathogenesis; however, these models cannot recapitulate all the aspects of this complex disorder as seen in each individual. Human pluripotent stem cells (hPSCs) can overcome the hurdles faced with the classical mouse models for studying IR. Human induced pluripotent stem cells (hiPSCs) can be generated from the somatic cells of the patients without the need to destroy a human embryo. Therefore, patient-specific hiPSCs can generate cells genetically identical to IR individuals, which can help in distinguishing between genetic and acquired defects in insulin sensitivity. Combining the technologies of genome editing and hiPSCs may provide important information about the genetic factors underlying the development of different forms of IR. Further studies are required to fill the gaps in understanding the pathogenesis of IR and diabetes. In this review, we summarize the factors involved in the development of IR in the insulin-target tissues leading to diabetes. Also, we highlight the use of hPSCs to understand the mechanisms underlying the development of IR.
Collapse
Affiliation(s)
- Ahmed K Elsayed
- Diabetes Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha 34110, Qatar
| | | | - Manjula Nandakumar
- Diabetes Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha 34110, Qatar
| | - Essam M Abdelalim
- Diabetes Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha 34110, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha 34110, Qatar
| |
Collapse
|
15
|
Sazonova MA, Sinyov VV, Ryzhkova AI, Sazonova MD, Kirichenko TV, Khotina VA, Khasanova ZB, Doroschuk NA, Karagodin VP, Orekhov AN, Sobenin IA. Some Molecular and Cellular Stress Mechanisms Associated with Neurodegenerative Diseases and Atherosclerosis. Int J Mol Sci 2021; 22:E699. [PMID: 33445687 PMCID: PMC7828120 DOI: 10.3390/ijms22020699] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 12/14/2022] Open
Abstract
Chronic stress is a combination of nonspecific adaptive reactions of the body to the influence of various adverse stress factors which disrupt its homeostasis, and it is also a corresponding state of the organism's nervous system (or the body in general). We hypothesized that chronic stress may be one of the causes occurence of several molecular and cellular types of stress. We analyzed literary sources and considered most of these types of stress in our review article. We examined genes and mutations of nuclear and mitochondrial genomes and also molecular variants which lead to various types of stress. The end result of chronic stress can be metabolic disturbance in humans and animals, leading to accumulation of reactive oxygen species (ROS), oxidative stress, energy deficiency in cells (due to a decrease in ATP synthesis) and mitochondrial dysfunction. These changes can last for the lifetime and lead to severe pathologies, including neurodegenerative diseases and atherosclerosis. The analysis of literature allowed us to conclude that under the influence of chronic stress, metabolism in the human body can be disrupted, mutations of the mitochondrial and nuclear genome and dysfunction of cells and their compartments can occur. As a result of these processes, oxidative, genotoxic, and cellular stress can occur. Therefore, chronic stress can be one of the causes forthe occurrence and development of neurodegenerative diseases and atherosclerosis. In particular, chronic stress can play a large role in the occurrence and development of oxidative, genotoxic, and cellular types of stress.
Collapse
Affiliation(s)
- Margarita A. Sazonova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia; (V.V.S.); (A.I.R.); (M.D.S.); (T.V.K.); (V.A.K.); (V.P.K.); (A.N.O.); (I.A.S.)
- Laboratory of Medical Genetics, National Medical Research Center of Cardiology, 121552 Moscow, Russia; (Z.B.K.); (N.A.D.)
| | - Vasily V. Sinyov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia; (V.V.S.); (A.I.R.); (M.D.S.); (T.V.K.); (V.A.K.); (V.P.K.); (A.N.O.); (I.A.S.)
- Laboratory of Medical Genetics, National Medical Research Center of Cardiology, 121552 Moscow, Russia; (Z.B.K.); (N.A.D.)
| | - Anastasia I. Ryzhkova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia; (V.V.S.); (A.I.R.); (M.D.S.); (T.V.K.); (V.A.K.); (V.P.K.); (A.N.O.); (I.A.S.)
| | - Marina D. Sazonova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia; (V.V.S.); (A.I.R.); (M.D.S.); (T.V.K.); (V.A.K.); (V.P.K.); (A.N.O.); (I.A.S.)
| | - Tatiana V. Kirichenko
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia; (V.V.S.); (A.I.R.); (M.D.S.); (T.V.K.); (V.A.K.); (V.P.K.); (A.N.O.); (I.A.S.)
- Laboratory of Medical Genetics, National Medical Research Center of Cardiology, 121552 Moscow, Russia; (Z.B.K.); (N.A.D.)
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Research Institute of Human Morphology, 117418 Moscow, Russia
| | - Victoria A. Khotina
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia; (V.V.S.); (A.I.R.); (M.D.S.); (T.V.K.); (V.A.K.); (V.P.K.); (A.N.O.); (I.A.S.)
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Research Institute of Human Morphology, 117418 Moscow, Russia
| | - Zukhra B. Khasanova
- Laboratory of Medical Genetics, National Medical Research Center of Cardiology, 121552 Moscow, Russia; (Z.B.K.); (N.A.D.)
| | - Natalya A. Doroschuk
- Laboratory of Medical Genetics, National Medical Research Center of Cardiology, 121552 Moscow, Russia; (Z.B.K.); (N.A.D.)
| | - Vasily P. Karagodin
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia; (V.V.S.); (A.I.R.); (M.D.S.); (T.V.K.); (V.A.K.); (V.P.K.); (A.N.O.); (I.A.S.)
- Department of Commodity Science and Expertise, Plekhanov Russian University of Economics, 125993 Moscow, Russia
| | - Alexander N. Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia; (V.V.S.); (A.I.R.); (M.D.S.); (T.V.K.); (V.A.K.); (V.P.K.); (A.N.O.); (I.A.S.)
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Research Institute of Human Morphology, 117418 Moscow, Russia
- Institute for Atherosclerosis Research, Skolkovo Innovative Centre, 143024 Moscow, Russia
| | - Igor A. Sobenin
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia; (V.V.S.); (A.I.R.); (M.D.S.); (T.V.K.); (V.A.K.); (V.P.K.); (A.N.O.); (I.A.S.)
- Laboratory of Medical Genetics, National Medical Research Center of Cardiology, 121552 Moscow, Russia; (Z.B.K.); (N.A.D.)
| |
Collapse
|
16
|
DhHP-6 ameliorates hepatic oxidative stress and insulin resistance in type 2 diabetes mellitus through the PI3K/AKT and AMPK pathway. Biochem J 2020; 477:2363-2381. [PMID: 32510127 DOI: 10.1042/bcj20200402] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 12/15/2022]
Abstract
Insulin resistance is one major features of type 2 diabetes mellitus (T2DM). Deuterohemin-βAla-His-Thr-Val-Glu-Lys (DhHP-6), a novel microperoxidase mimetic designed and synthesized based on microperoxidase 11 (MP-11), can scavenge reactive oxygen species (ROS) in vivo. In our previous studies, we showed that oral DhHP-6 could reduce blood glucose and improve insulin resistance. To investigate the mechanisms of how DhHP-6 ameliorates oxidative stress and insulin resistance, we established T2DM mouse models and glucosamine-induced HepG2 cell insulin resistance models. The results suggested that DhHP-6 decreased blood glucose, increased antioxidant enzyme activity, and inhibited glycogen synthesis in T2DM mice. In addition, DhHP-6 improved insulin resistance by activating phosphatidylinositol 3-kinase (PI3K)/AKT, and AMP-activated protein kinase (AMPK) pathway in T2DM mice. Furthermore, DhHP-6 also activated PI3K/AKT and AMPK pathway in glucosamine-induced HepG2 cells. However, LY294002 did not completely inhibit AKT phosphorylation, and partially inhibited AMPK phosphorylation, whilst compound C only partially reduced AMPK phosphorylation, and also partially inhibited AKT phosphorylation, suggesting that AKT and AMPK interact to improve insulin resistance. Thus, these data suggest that DhHP-6 attenuates insulin resistance via the PI3K/AKT and AMPK pathway.
Collapse
|
17
|
Developmental Programming and Glucolipotoxicity: Insights on Beta Cell Inflammation and Diabetes. Metabolites 2020; 10:metabo10110444. [PMID: 33158303 PMCID: PMC7694373 DOI: 10.3390/metabo10110444] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/23/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
Stimuli or insults during critical developmental transitions induce alterations in progeny anatomy, physiology, and metabolism that may be transient, sometimes reversible, but often durable, which defines programming. Glucolipotoxicity is the combined, synergistic, deleterious effect of simultaneously elevated glucose (chronic hyperglycemia) and saturated fatty acids (derived from high-fat diet overconsumption and subsequent metabolism) that are harmful to organs, micro-organs, and cells. Glucolipotoxicity induces beta cell death, dysfunction, and failure through endoplasmic reticulum and oxidative stress and inflammation. In beta cells, the misfolding of pro/insulin proteins beyond the cellular threshold triggers the unfolded protein response and endoplasmic reticulum stress. Consequentially there is incomplete and inadequate pro/insulin biosynthesis and impaired insulin secretion. Cellular stress triggers cellular inflammation, where immune cells migrate to, infiltrate, and amplify in beta cells, leading to beta cell inflammation. Endoplasmic reticulum stress reciprocally induces beta cell inflammation, whereas beta cell inflammation can self-activate and further exacerbate its inflammation. These metabolic sequelae reflect the vicious cycle of beta cell stress and inflammation in the pathophysiology of diabetes.
Collapse
|
18
|
Ke C, Stukel TA, Shah BR, Lau E, Ma RC, So WY, Kong AP, Chow E, Chan JCN, Luk A. Age at diagnosis, glycemic trajectories, and responses to oral glucose-lowering drugs in type 2 diabetes in Hong Kong: A population-based observational study. PLoS Med 2020; 17:e1003316. [PMID: 32946450 PMCID: PMC7500681 DOI: 10.1371/journal.pmed.1003316] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/14/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Lifetime glycemic exposure and its relationship with age at diagnosis in type 2 diabetes (T2D) are unknown. Pharmacologic glycemic management strategies for young-onset T2D (age at diagnosis <40 years) are poorly defined. We studied how age at diagnosis affects glycemic exposure, glycemic deterioration, and responses to oral glucose-lowering drugs (OGLDs). METHODS AND FINDINGS In a population-based cohort (n = 328,199; 47.2% women; mean age 34.6 and 59.3 years, respectively, for young-onset and usual-onset [age at diagnosis ≥40 years] T2D; 2002-2016), we used linear mixed-effects models to estimate the association between age at diagnosis and A1C slope (glycemic deterioration) and tested for an interaction between age at diagnosis and responses to various combinations of OGLDs during the first decade after diagnosis. In a register-based cohort (n = 21,016; 47.1% women; mean age 43.8 and 58.9 years, respectively, for young- and usual-onset T2D; 2000-2015), we estimated the glycemic exposure from diagnosis until age 75 years. People with young-onset T2D had a higher mean A1C (8.0% [standard deviation 0.15%]) versus usual-onset T2D (7.6% [0.03%]) throughout the life span (p < 0.001). The cumulative glycemic exposure was >3 times higher for young-onset versus usual-onset T2D (41.0 [95% confidence interval 39.1-42.8] versus 12.1 [11.8-12.3] A1C-years [1 A1C-year = 1 year with 8% average A1C]). Younger age at diagnosis was associated with faster glycemic deterioration (A1C slope over time +0.08% [0.078-0.084%] per year for age at diagnosis 20 years versus +0.02% [0.016-0.018%] per year for age at diagnosis 50 years; p-value for interaction <0.001). Age at diagnosis ≥60 years was associated with glycemic improvement (-0.004% [-0.005 to -0.004%] and -0.02% [-0.027 to -0.0244%] per year for ages 60 and 70 years at diagnosis, respectively; p-value for interaction <0.001). Responses to OGLDs differed by age at diagnosis (p-value for interaction <0.001). Those with young-onset T2D had smaller A1C decrements for metformin-based combinations versus usual-onset T2D (metformin alone: young-onset -0.15% [-0.105 to -0.080%], usual-onset -0.17% [-0.179 to -0.169%]; metformin, sulfonylurea, and dipeptidyl peptidase-4 inhibitor: young-onset -0.44% [-0.476 to -0.405%], usual-onset -0.48% [-0.498 to -0.459%]; metformin and α-glucosidase inhibitor: young-onset -0.40% [-0.660 to -0.144%], usual-onset -0.25% [-0.420 to -0.077%]) but greater responses to other combinations containing sulfonylureas (sulfonylurea alone: young-onset -0.08% [-0.099 to -0.065%], usual-onset +0.06% [+0.059 to +0.072%]; sulfonylurea and α-glucosidase inhibitor: young-onset -0.10% [-0.266 to 0.064%], usual-onset: 0.25% [+0.196% to +0.312%]). Limitations include possible residual confounding and unknown generalizability outside Hong Kong. CONCLUSIONS In this study, we observed excess glycemic exposure and rapid glycemic deterioration in young-onset T2D, indicating that improved treatment strategies are needed in this setting. The differential responses to OGLDs between young- and usual-onset T2D suggest that better disease classification could guide personalized therapy.
Collapse
Affiliation(s)
- Calvin Ke
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
- Department of Medicine, University of Toronto, Canada
| | - Thérèse A. Stukel
- Institute of Health Policy, Management and Evaluation, University of Toronto, Canada
- ICES, Toronto, Canada
| | - Baiju R. Shah
- Department of Medicine, University of Toronto, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Canada
- ICES, Toronto, Canada
- Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Eric Lau
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
- Asia Diabetes Foundation, Metropole Square, Shatin, Hong Kong SAR, China
| | - Ronald C. Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
- Hong Kong Institute of Diabetes and Obesity and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Wing-Yee So
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Alice P. Kong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
- Hong Kong Institute of Diabetes and Obesity and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Elaine Chow
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Juliana C. N. Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
- Asia Diabetes Foundation, Metropole Square, Shatin, Hong Kong SAR, China
- Hong Kong Institute of Diabetes and Obesity and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
- * E-mail:
| | - Andrea Luk
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
- Asia Diabetes Foundation, Metropole Square, Shatin, Hong Kong SAR, China
- Hong Kong Institute of Diabetes and Obesity and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| |
Collapse
|
19
|
Said E, Mousa S, Fawzi M, Sabry NA, Farid S. Combined effect of high-dose vitamin A, vitamin E supplementation, and zinc on adult patients with diabetes: A randomized trial. J Adv Res 2020; 28:27-33. [PMID: 33364042 PMCID: PMC7753230 DOI: 10.1016/j.jare.2020.06.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 12/24/2022] Open
Abstract
In type 2 diabetes mellitus (T2DM), hyperglycemia leads to oxidative insult. Vitamins A and E have antioxidant potentials and may help in managing diabetes. The combined effect of high-dose vitamin A plus E supplementation with and without zinc on T2DM, has never been examined. Thus, this study aimed to evaluate and compare the effect of high-dose vitamin A plus E supplementation (AE) versus high-dose vitamin A plus E with zinc (AEZ), on different diabetic parameters. Ninety-eight patients with T2DM were randomized to receive either: 50,000 IU vitamin A and 100 mg vitamin E (AE group, N = 36), an equivalent dose of vitamin A and E combined with 25 mg zinc (AEZ group, N = 35), or no supplements (control group, N = 27) for three months. Compared to control, AEZ group showed significant reductions in fasting blood glucose, 2 h postprandial blood glucose, and glycated hemoglobin (HbA1c) with significant increases in homeostasis model assessment of beta-cell function and difference value of fasting insulin. Two hair loss cases were recorded in both treated groups. Although vitamin A needs dose moderation, these results suggest that, high-dose vitamin A plus E supplementation combined with zinc may improve glycemic control, β-cell function, and insulin secretion in adults with T2DM.
Collapse
Affiliation(s)
- Eman Said
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Shrook Mousa
- Department of Internal Medicine, Kasr Alainy Faculty of Medicine, Cairo University, Cairo 11562, Egypt
| | - May Fawzi
- Department of Internal Medicine, Kasr Alainy Faculty of Medicine, Cairo University, Cairo 11562, Egypt
| | - Nirmeen A Sabry
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Samar Farid
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
20
|
AL-Megrin WA, El-Khadragy MF, Hussein MH, Mahgoub S, Abdel-Mohsen DM, Taha H, Bakkar AAA, Abdel Moneim AE, Amin HK. Green Coffea arabica Extract Ameliorates Testicular Injury in High-Fat Diet/Streptozotocin-Induced Diabetes in Rats. J Diabetes Res 2020; 2020:6762709. [PMID: 32626781 PMCID: PMC7306074 DOI: 10.1155/2020/6762709] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 04/25/2020] [Indexed: 12/14/2022] Open
Abstract
Diabetes mellitus (DM) is a chronic endocrine disease characterized by persistent hyperglycemia. Oxidative damage, inflammatory cytokines, and apoptotic cell death play a major role in the induction and progression of male testicular damage. Plant-derived phytochemicals such as green coffee (Coffea arabica) can possess antidiabetic effects with little toxicity. The current study is aimed at investigating the therapeutic roles of green coffee in diabetic testicular injury stimulated by high-fat diet/streptozotocin administration. Diabetes mellitus was induced by a high-fat diet and a single dose of streptozotocin (STZ) (35 mg kg-1) in male albino rats. Diabetic animals were orally given two different concentrations of green coffee (50 mg kg-1 and 100 mg kg-1) for 28 days. The levels of testosterone, luteinizing hormone, and follicle-stimulating hormone and parameters of oxidative stress, inflammation, and apoptosis were measured. mRNAs and protein levels were detected quantitatively by real-time PCR and ELISA, respectively. In the diabetic group, the levels of testosterone, luteinizing hormone, and follicle-stimulating hormone showed a significant reduction while they increased significantly after green coffee treatment. A significant increase of antioxidant markers glutathione, superoxide dismutase, catalase, glutathione peroxidase, and glutathione reductase along with decreased levels of lipid peroxides and nitric oxide was observed after green coffee treatment in the diabetic group. Finally, the levels of IL-1β, TNF-α, Bax, and caspase-3 were also decreased in both treated groups (metformin and green coffee) when compared to the diabetic group. We conclude that testicular oxidative impairment induced by a high-fat diet (HFD) and STZ can be reversed by green coffee. Administration of green coffee could represent a promising therapeutic agent which can help the treatment of type 2 DM-induced testicular dysfunction.
Collapse
Affiliation(s)
- Wafa A. AL-Megrin
- Biology Department, Faculty of Science, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - Manal F. El-Khadragy
- Biology Department, Faculty of Science, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Manal H. Hussein
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Shahenda Mahgoub
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Doaa M. Abdel-Mohsen
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Heba Taha
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Ashraf A. A. Bakkar
- Faculty of Biotechnology, Modern Sciences and Arts University (MSA), Giza, Egypt
| | - Ahmed E. Abdel Moneim
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Hatem K. Amin
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| |
Collapse
|
21
|
Cripps MJ, Bagnati M, Jones TA, Ogunkolade BW, Sayers SR, Caton PW, Hanna K, Billacura MP, Fair K, Nelson C, Lowe R, Hitman GA, Berry MD, Turner MD. Identification of a subset of trace amine-associated receptors and ligands as potential modulators of insulin secretion. Biochem Pharmacol 2020; 171:113685. [DOI: 10.1016/j.bcp.2019.113685] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/24/2019] [Indexed: 12/19/2022]
|
22
|
Quiclet C, Dittberner N, Gässler A, Stadion M, Gerst F, Helms A, Baumeier C, Schulz TJ, Schürmann A. Pancreatic adipocytes mediate hypersecretion of insulin in diabetes-susceptible mice. Metabolism 2019; 97:9-17. [PMID: 31108105 DOI: 10.1016/j.metabol.2019.05.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/26/2019] [Accepted: 05/13/2019] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Ectopic fat accumulation in the pancreas in response to obesity and its implication on the onset of type 2 diabetes remain poorly understood. Intermittent fasting (IF) is known to improve glucose homeostasis and insulinresistance. However, the effects of IF on fat in the pancreas and β-cell function remain largely unknown. Our aim was to evaluate the impact of IF on pancreatic fat accumulation and its effects on islet function. METHODS New Zealand Obese (NZO) mice were fed a high-fat diet ad libitum (NZO-AL) or fasted every other day (intermittent fasting, NZO-IF) and pancreatic fat accumulation, glucose homoeostasis, insulin sensitivity, and islet function were determined and compared to ad libitum-fed B6.V-Lepob/ob (ob/ob) mice. To investigate the crosstalk of pancreatic adipocytes and islets, co-culture experiments were performed. RESULTS NZO-IF mice displayed better glucose homeostasis and lower fat accumulation in both the pancreas (-32%) and the liver (-35%) than NZO-AL mice. Ob/ob animals were insulin-resistant and had low fat in the pancreas but high fat in the liver. NZO-AL mice showed increased fat accumulation in both organs and exhibited an impaired islet function. Co-culture experiments demonstrated that pancreatic adipocytes induced a hypersecretion of insulin and released higher levels of free fatty acids than adipocytes of inguinal white adipose tissue. CONCLUSIONS These results suggest that pancreatic fat participates in diabetes development, but can be prevented byIF.
Collapse
Affiliation(s)
- Charline Quiclet
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg 85764, Germany
| | - Nicole Dittberner
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany.
| | - Anneke Gässler
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg 85764, Germany
| | - Mandy Stadion
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg 85764, Germany.
| | - Felicia Gerst
- German Center for Diabetes Research (DZD), München-Neuherberg 85764, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard Karls University of Tübingen, 72076, Tübingen, Germany.
| | - Anett Helms
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg 85764, Germany.
| | - Christian Baumeier
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg 85764, Germany
| | - Tim J Schulz
- German Center for Diabetes Research (DZD), München-Neuherberg 85764, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard Karls University of Tübingen, 72076, Tübingen, Germany; Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany.
| | - Annette Schürmann
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg 85764, Germany; Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany.
| |
Collapse
|
23
|
Lee T, Yun S, Jeong JH, Jung TW. Asprosin impairs insulin secretion in response to glucose and viability through TLR4/JNK-mediated inflammation. Mol Cell Endocrinol 2019; 486:96-104. [PMID: 30853600 DOI: 10.1016/j.mce.2019.03.001] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 03/02/2019] [Accepted: 03/04/2019] [Indexed: 01/02/2023]
Abstract
Severe inflammation in the islets is observed in obese patients with type 2 diabetes. Inflammation in the islets is caused by obesity-induced serum free fatty acids. Asprosin is a fasting-induced adipokine, which contributes to hepatic glucose production. However, the effects of asprosin on inflammation and cellular dysfunction in pancreatic β-cells remain to be elucidated. Here, we demonstrated that treatment of mouse insulinoma MIN6 cells and human primary islets containing β-cells with palmitate increased asprosin expression and secretion. Treatment of MIN6 cells and human primary islets with palmitate increased phosphorylation of the inflammatory marker nuclear factor-kappa B (NFκB) and the release of pro-inflammatory cytokines including TNF and MCP-1 and decreased glucose-stimulated insulin secretion and cell viability. However, siRNA-mediated suppression of asprosin reversed these changes. Recombinant asprosin treatment of MIN6 cells and human primary islets augmented the inflammation response, cellular dysfunction, and apoptosis in a dose-dependent manner. Asprosin induced toll-like receptor (TLR) 4 expression and JNK phosphorylation. siRNA for TLR4 or JNK mitigated the effects of asprosin on inflammation and cellular dysfunction. These results suggest that palmitate-derived asprosin secretion from β-cells results in their inflammation and dysfunction through a TLR4/JNK-mediated pathway. This report suggests asprosin as a novel therapeutic target for the treatment of type 2 diabetes through preservation of β-cell function.
Collapse
Affiliation(s)
- Taeseung Lee
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, South Korea; Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Subin Yun
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, South Korea
| | - Tae Woo Jung
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, South Korea.
| |
Collapse
|
24
|
Graus-Nunes F, Souza-Mello V. The renin-angiotensin system as a target to solve the riddle of endocrine pancreas homeostasis. Biomed Pharmacother 2018; 109:639-645. [PMID: 30404071 DOI: 10.1016/j.biopha.2018.10.191] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 10/27/2022] Open
Abstract
Local renin-angiotensin system (RAS) in the pancreas is linked to the modulation of glucose-stimulated insulin secretion (GSIS) in beta cells and insulin sensitivity in target tissues, emerging as a promising tool in the prevention and/or treatment of obesity, diabetes, and systemic arterial hypertension. Insulin resistance alters pancreatic islet cell distribution and morphology and hypertrophied islets exhibit upregulated angiotensin II type 1 receptor, which drives oxidative stress, apoptosis, and fibrosis, configuring beta cell dysfunction and diminishing islet lifespan. Pharmacological modulation of RAS has shown beneficial effects in diet-induced obesity model, mainly related to the translational potential that angiotensin receptor blockers and ECA2/ANG (1-7)/MAS receptor axis modulation have when it comes to islet preservation and type 2 diabetes prevention and/or treatment. This review describes the existing evidence for different approaches to blocking RAS elements in the management of insulin resistance and diabetes and focuses on islet remodeling and GSIS in rodents and humans.
Collapse
Affiliation(s)
- Francielle Graus-Nunes
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil.
| |
Collapse
|
25
|
Huang Q, You W, Li Y, Sun Y, Zhou Y, Zhang Y, Liu D, Zhan S, Zhu Y, Han X. Glucolipotoxicity-Inhibited miR-299-5p Regulates Pancreatic β-Cell Function and Survival. Diabetes 2018; 67:2280-2292. [PMID: 30131392 DOI: 10.2337/db18-0223] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 08/07/2018] [Indexed: 11/13/2022]
Abstract
Inhibition of microRNAs (miRNAs) essential for pancreatic β-cell biology (e.g., miR-375) results in β-cell failure and diabetes in rodent models. Whether the downregulation of miRNAs in pancreatic islets is involved in the development of human type 2 diabetes remains unclear. Here, with the use of an miRNA microarray, we identified a set of miRNAs that were differentially expressed in healthy human islets under glucolipotoxic conditions. A downregulated miRNA, miR-299-5p, was preferentially studied because its inhibition causes dramatic β-cell dysfunction and apoptosis. Proteomic profiling and bioinformatics methods identified four target genes, including a Trp53 effector, Perp, that were further confirmed by luciferase reporter assays. We narrowed down the effector of miR-299-5p downregulation to PERP owing to its upregulation in islets from diabetic rodents. Indeed, Perp inhibition prevented the β-cell impairment caused by either miR-299-5p reduction or glucolipotoxicity. Additional investigations confirmed the modulatory effect of PERP on insulin secretion. Collectively, miR-299-5p appears to be an essential regulator of β-cell biology, and its downregulation links PERP enhancement to β-cell dysfunction and apoptosis in glucolipotoxic settings. Our work demonstrates a novel mechanism of glucolipotoxicity-induced β-cell failure mediated through miR-299-5p downregulation.
Collapse
Affiliation(s)
- Qiqing Huang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Geriatrics of Jiangsu Province, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weiyan You
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory for Neurodegenerative Disease of Jiangsu Province, Department of Neurobiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yating Li
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yi Sun
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuncai Zhou
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yan Zhang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dechen Liu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shanshan Zhan
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of General Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yunxia Zhu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
26
|
Hong SW, Lee J, Cho JH, Kwon H, Park SE, Rhee EJ, Park CY, Oh KW, Park SW, Lee WY. Pioglitazone Attenuates Palmitate-Induced Inflammation and Endoplasmic Reticulum Stress in Pancreatic β-Cells. Endocrinol Metab (Seoul) 2018; 33:105-113. [PMID: 29589392 PMCID: PMC5874186 DOI: 10.3803/enm.2018.33.1.105] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The nuclear receptor peroxisome proliferator-activator gamma (PPARγ) is a useful therapeutic target for obesity and diabetes, but its role in protecting β-cell function and viability is unclear. METHODS To identify the potential functions of PPARγ in β-cells, we treated mouse insulinoma 6 (MIN6) cells with the PPARγ agonist pioglitazone in conditions of lipotoxicity, endoplasmic reticulum (ER) stress, and inflammation. RESULTS Palmitate-treated cells incubated with pioglitazone exhibited significant improvements in glucose-stimulated insulin secretion and the repression of apoptosis, as shown by decreased caspase-3 cleavage and poly (adenosine diphosphate [ADP]-ribose) polymerase activity. Pioglitazone also reversed the palmitate-induced expression of inflammatory cytokines (tumor necrosis factor α, interleukin 6 [IL-6], and IL-1β) and ER stress markers (phosphor-eukaryotic translation initiation factor 2α, glucose-regulated protein 78 [GRP78], cleaved-activating transcription factor 6 [ATF6], and C/EBP homologous protein [CHOP]), and pioglitazone significantly attenuated inflammation and ER stress in lipopolysaccharide- or tunicamycin-treated MIN6 cells. The protective effect of pioglitazone was also tested in pancreatic islets from high-fat-fed KK-Ay mice administered 0.02% (wt/wt) pioglitazone or vehicle for 6 weeks. Pioglitazone remarkably reduced the expression of ATF6α, GRP78, and monocyte chemoattractant protein-1, prevented α-cell infiltration into the pancreatic islets, and upregulated glucose transporter 2 (Glut2) expression in β-cells. Moreover, the preservation of β-cells by pioglitazone was accompanied by a significant reduction of blood glucose levels. CONCLUSION Altogether, these results support the proposal that PPARγ agonists not only suppress insulin resistance, but also prevent β-cell impairment via protection against ER stress and inflammation. The activation of PPARγ might be a new therapeutic approach for improving β-cell survival and insulin secretion in patients with diabetes mellitus.
Collapse
Affiliation(s)
- Seok Woo Hong
- Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jinmi Lee
- Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jung Hwan Cho
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyemi Kwon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Se Eun Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Eun Jung Rhee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Cheol Young Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ki Won Oh
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sung Woo Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Won Young Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
27
|
Sadri H, Larki NN, Kolahian S. Hypoglycemic and Hypolipidemic Effects of Leucine, Zinc, and Chromium, Alone and in Combination, in Rats with Type 2 Diabetes. Biol Trace Elem Res 2017; 180:246-254. [PMID: 28409409 DOI: 10.1007/s12011-017-1014-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/04/2017] [Indexed: 01/08/2023]
Abstract
For the increasing development of diabetes, dietary habits and using appropriate supplements can play important roles in the treatment or reduction of risk for this disease. The objective of this study was to investigate the effects of leucine (Leu), zinc (Zn), and chromium (Cr) supplementation, alone or in combination, in rats with type 2 diabetes (T2D). Seventy-seven adult male Wistar rats were randomly assigned in 11 groups, using nutritional supplements and insulin (INS) or glibenclamide (GLC). Supplementing Leu significantly reduced blood glucose, triglycerides (TG), nonesterified fatty acids (NEFA), low-density lipoprotein (LDL), and increased high-density lipoprotein (HDL) concentrations compared to vehicle-treated T2D animals, and those improvements were associated with reduced area under the 2-h blood glucose response curve (AUC). Supplementation of T2D animals with Zn improved serum lipid profile as well as blood glucose concentrations but was not comparable with the INS, GLC, and Leu groups. Supplementary Cr did not improve blood glucose and AUC in T2D rats, whereas it reduced serum TG and LDL and increased HDL concentrations. In conclusion, supplementation of diabetic rats with Leu was more effective in improving blood glucose and consequently decreasing glucose AUC than other nutritional supplements. Supplementary Zn and Cr only improved serum lipid profile. The combination of the nutritional supplements did not improve blood glucose level. Nevertheless, supplementation with Leu-Zn, Leu-Cr, Zn-Cr, and Leu-Zn-Cr led to an improved response in serum lipid profile over each supplement given alone.
Collapse
Affiliation(s)
- Hassan Sadri
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, 516616471, Iran.
| | - Negar Nowroozi Larki
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology and ICePhA, University of Tuebingen, 72074, Tuebingen, Germany
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, 516616471, Iran
| | - Saeed Kolahian
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology and ICePhA, University of Tuebingen, 72074, Tuebingen, Germany
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, 516616471, Iran
| |
Collapse
|
28
|
Jung C, Lichtenauer M, Strodthoff D, Winkels H, Wernly B, Bürger C, Kamchybekov U, Lutgens E, Figulla HR, Gerdes N. Alterations in systemic levels of Th1, Th2, and Th17 cytokines in overweight adolescents and obese mice. Pediatr Diabetes 2017; 18:714-721. [PMID: 27597513 DOI: 10.1111/pedi.12435] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 07/18/2016] [Accepted: 08/03/2016] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Obesity represents a major problem for patients and health care systems in most industrialized countries. A chronic inflammatory state in obese individuals leads to disease conditions associated with activation of cellular immune mechanisms. Here, we sought to investigate the role of Th1-, Th2-, and Th17-related cytokines in overweight adolescents and mice on a high-fat diet. METHODS Plasma samples were obtained from 79 male adolescents aged 13-17 years. Thirty-seven of them had a body mass index (BMI) above the 90th age-specific percentile. Th1, Th2, and Th17 cytokines were measured using Bio-Plex multiplex technology (Bio-Rad, Hercules, USA). In an experimental approach, mice were fed with high-fat (HFD) or normal chow for 15 weeks. RESULTS Interleukin (IL)-17 concentrations were significantly decreased in overweight adolescents compared to lean controls [99.8 ± 7.3 pg/mL standard error of the mean (SEM) vs 146.6 ± 11.5 pg/mL SEM P = .001]. Levels of IL-17 correlated significantly with anthropometrical parameters of obesity. A concordant response was found in mice consuming a HFD for 15 weeks compared to controls (861 ± 165 pg/mL SEM vs 1575 ± 187 pg/ml SEM, P = .0183). However, a biphasic response was evident for most Th1, Th2, and Th17 cytokines as levels initially increased within the first 5 weeks on HFD and showed a decline afterwards. CONCLUSIONS In contrast to previous studies showing elevated levels of IL-17 in obese adults, we found a decreasing trend in overweight adolescents. This difference could possibly be related to the fact that disease conditions associated with obesity such as hypertension, vascular pathologies, diabetes, and a triggering of the Th1/Th17 axis were not yet present in overweight teenagers.
Collapse
Affiliation(s)
- Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Michael Lichtenauer
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Daniela Strodthoff
- Department of Medicine, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Holger Winkels
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, München, Germany
| | - Bernhard Wernly
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Christina Bürger
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, München, Germany
| | - Uran Kamchybekov
- Clinic of Internal Medicine I, Friedrich-Schiller-University, Jena, Germany
| | - Esther Lutgens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, München, Germany.,Department of Medical Biochemistry, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | | | - Norbert Gerdes
- Department of Medicine, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden.,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, München, Germany.,Department of Medical Biochemistry, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
29
|
Freitas HR, Ferreira GDC, Trevenzoli IH, Oliveira KDJ, de Melo Reis RA. Fatty Acids, Antioxidants and Physical Activity in Brain Aging. Nutrients 2017; 9:nu9111263. [PMID: 29156608 PMCID: PMC5707735 DOI: 10.3390/nu9111263] [Citation(s) in RCA: 40] [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: 10/01/2017] [Revised: 11/11/2017] [Accepted: 11/14/2017] [Indexed: 12/25/2022] Open
Abstract
Polyunsaturated fatty acids and antioxidants are important mediators in the central nervous system. Lipid derivatives may control the production of proinflammatory agents and regulate NF-κB activity, microglial activation, and fatty acid oxidation; on the other hand, antioxidants, such as glutathione and ascorbate, have been shown to signal through transmitter receptors and protect against acute and chronic oxidative stress, modulating the activity of different signaling pathways. Several authors have investigated the role of these nutrients in the brains of the young and the aged in degenerative diseases such as Alzheimer’s and Parkinson’s, and during brain aging due to adiposity- and physical inactivity-mediated metabolic disturbances, chronic inflammation, and oxidative stress. Through a literature review, we aimed to highlight recent data on the role of adiposity, fatty acids, antioxidants, and physical inactivity in the pathophysiology of the brain and in the molecular mechanisms of senescence. Data indicate the complexity and necessity of endogenous/dietary antioxidants for the maintenance of redox status and the control of neuroglial signaling under stress. Recent studies also indicate that omega-3 and -6 fatty acids act in a competitive manner to generate mediators for energy metabolism, influencing feeding behavior, neural plasticity, and memory during aging. Finding pharmacological or dietary resources that mitigate or prevent neurodegenerative affections continues to be a great challenge and requires additional effort from researchers, clinicians, and nutritionists in the field.
Collapse
Affiliation(s)
- Hércules Rezende Freitas
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil.
| | - Gustavo da Costa Ferreira
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil.
- Laboratory of Neuroenergetics and Inborn Errors of Metabolism, Institute of Medical Biochemistry Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil.
| | - Isis Hara Trevenzoli
- Laboratory of Molecular Endocrinology, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil.
| | - Karen de Jesus Oliveira
- Laboratory of Endocrine Physiology and Metabology, Biomedical Institute, Universidade Federal Fluminense, Niterói 24210-130, Brazil.
| | - Ricardo Augusto de Melo Reis
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil.
| |
Collapse
|
30
|
Sodium fluorocitrate having protective effect on palmitate-induced beta cell death improves hyperglycemia in diabetic db/db mice. Sci Rep 2017; 7:12916. [PMID: 29018279 PMCID: PMC5635019 DOI: 10.1038/s41598-017-13365-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 09/21/2017] [Indexed: 02/06/2023] Open
Abstract
Beta cell loss and insulin resistance play roles in the pathogenesis of type 2 diabetes. Elevated levels of free fatty acids in plasma might contribute to the loss of beta cells. The objective of this study was to find a chemical that could protect against palmitate-induced beta cell death and investigate whether such chemical could improve hyperglycemia in mouse model of type 2 diabetes. Sodium fluorocitrate (SFC), an aconitase inhibitor, was found to be strongly and specifically protective against palmitate-induced INS-1 beta cell death. However, the protective effect of SFC on palmitate-induced cell death was not likely to be due to its inhibitory activity for aconitase since inhibition or knockdown of aconitase failed to protect against palmitate-induced cell death. Since SFC inhibited the uptake of palmitate into INS-1 cells, reduced metabolism of fatty acids was thought to be involved in SFC’s protective effect. Ten weeks of treatment with SFC in db/db diabetic mice reduced glucose level but remarkably increased insulin level in the plasma. SFC improved impairment of glucose-stimulated insulin release and also reduced the loss of beta cells in db/db mice. Conclusively, SFC possessed protective effect against palmitate-induced lipotoxicity and improved hyperglycemia in mouse model of type 2 diabetes.
Collapse
|
31
|
Phenolic constituents and modulatory effects of Raffia palm leaf ( Raphia hookeri) extract on carbohydrate hydrolyzing enzymes linked to type-2 diabetes. J Tradit Complement Med 2017; 7:494-500. [PMID: 29034198 PMCID: PMC5634752 DOI: 10.1016/j.jtcme.2017.01.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 08/09/2016] [Accepted: 01/12/2017] [Indexed: 01/05/2023] Open
Abstract
This study sought to investigate the effects of Raffia palm (Raphia hookeri) leaf extract on enzymes linked to type-2 diabetes mellitus (T2DM) and pro-oxidant induced oxidative stress in rat pancreas. The extract was prepared and its α-amylase and α-glucosidase inhibitory effects were determined. Radical [2,2-diphenyl-1-picrylhydrazyl (DPPH)] scavenging and Fe2+-chelating abilities, and inhibition of Fe2+-induced lipid peroxidation in rat pancreas homogenate were assessed. Furthermore, total phenol and flavonoid contents, reducing property, and high performance liquid chromatography diode array detector (HPLC-DAD) fingerprint of the extract were also determined. Our results revealed that the extract inhibited α-amylase (IC50 = 110.4 μg/mL) and α-glucosidase (IC50 = 99.96 μg/mL) activities in concentration dependent manners which were lower to the effect of acarbose (amylase: IC50 = 18.30 μg/mL; glucosidase: IC50 = 20.31 μg/mL). The extract also scavenged DPPH radical, chelated Fe2+ and inhibited Fe2+-induced lipid peroxidation in rat pancreas all in concentration dependent manners with IC50 values of 402.9 μg/mL, 108.9 μg/mL and 367.0 μg/mL respectively. The total phenol and flavonoid contents were 39.73 mg GAE/g and 21.88 mg QAE/g respectively, while the reducing property was 25.62 mg AAE/g. The HPLC analysis revealed the presence of chlorogenic acid (4.17 mg/g) and rutin (5.11 mg/g) as the major phenolic compounds in the extract. Therefore, the ability of the extract to inhibit carbohydrate hydrolyzing enzymes and protect against pancreatic oxidative damage may be an important mechanisms supporting its antidiabetic properties and could make Raffia palm leaf useful in complementary/alternative therapy for management of T2DM. However, further studies such as in vivo should be carried out.
Collapse
|
32
|
Diz-Chaves Y, Gil-Lozano M, Toba L, Fandiño J, Ogando H, González-Matías LC, Mallo F. Stressing diabetes? The hidden links between insulinotropic peptides and the HPA axis. J Endocrinol 2016; 230:R77-94. [PMID: 27325244 DOI: 10.1530/joe-16-0118] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 06/20/2016] [Indexed: 12/25/2022]
Abstract
Diabetes mellitus exerts metabolic stress on cells and it provokes a chronic increase in the long-term activity of the hypothalamus-pituitary-adrenocortical (HPA) axis, perhaps thereby contributing to insulin resistance. GLP-1 receptor (GLP-1R) agonists are pleiotropic hormones that not only affect glycaemic and metabolic control, but they also produce many other effects including activation of the HPA axis. In fact, several of the most relevant effects of GLP-1 might involve, at least in part, the modulation of the HPA axis. Thus, the anorectic activity of GLP-1 could be mediated by increasing CRF at the hypothalamic level, while its lipolytic effects could imply a local increase in glucocorticoids and glucocorticoid receptor (GC-R) expression in adipose tissue. Indeed, the potent activation of the HPA axis by GLP-1R agonists occurs within the range of therapeutic doses and with a short latency. Interestingly, the interactions of GLP-1 with the HPA axis may underlie most of the effects of GLP-1 on food intake control, glycaemic metabolism, adipose tissue biology and the responses to stress. Moreover, such activity has been observed in animal models (mice and rats), as well as in normal humans and in type I or type II diabetic patients. Accordingly, better understanding of how GLP-1R agonists modulate the activity of the HPA axis in diabetic subjects, especially obese individuals, will be crucial to design new and more efficient therapies for these patients.
Collapse
Affiliation(s)
- Yolanda Diz-Chaves
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Manuel Gil-Lozano
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Laura Toba
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Juan Fandiño
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Hugo Ogando
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Lucas C González-Matías
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Federico Mallo
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| |
Collapse
|
33
|
Cheng SH, Ismail A, Anthony J, Ng OC, Hamid AA, Yusof BNM. Effect of Cosmos caudatus (Ulam raja) supplementation in patients with type 2 diabetes: Study protocol for a randomized controlled trial. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 16:84. [PMID: 26920910 PMCID: PMC4769500 DOI: 10.1186/s12906-016-1047-7] [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] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 02/12/2016] [Indexed: 11/21/2022]
Abstract
BACKGROUND Type 2 diabetes mellitus is a major health threat worldwide. Cosmos caudatus is one of the medicinal plants used to treat type 2 diabetes. Therefore, this study aims to determine the effectiveness and safety of C. caudatus in patients with type 2 diabetes. Metabolomic approach will be carried out to compare the metabolite profiles between C. Caudatus treated diabetic patients and diabetic controls. METHODS AND DESIGN This is a single-center, randomized, controlled, two-arm parallel design clinical trial that will be carried out in a tertiary hospital in Malaysia. In this study, 100 patients diagnosed with type 2 diabetes will be enrolled. Diabetic patients who meet the eligibility criteria will be randomly allocated to two groups, which are diabetic C. caudatus treated(U) group and diabetic control (C) group. Primary and secondary outcomes will be measured at baseline, 4, 8, and 12 weeks. The serum and urine metabolome of both groups will be examined using proton NMR spectroscopy. DISCUSSION The study will be the first randomized controlled trial to assess whether C. caudatus can confer beneficial effect in patients with type 2 diabetes. The results of this trial will provide clinical evidence on the effectiveness and safety of C. caudatus in patients with type 2 diabetes. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT02322268.
Collapse
Affiliation(s)
- Shi-Hui Cheng
- Department of Nutrition and Dietetics, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| | - Amin Ismail
- Department of Nutrition and Dietetics, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Research Centre of Excellent for Nutrition and Non-communicable Diseases, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43300, Selangor, Malaysia
| | - Joseph Anthony
- Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43300, Selangor, Malaysia
| | - Ooi Chuan Ng
- Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43300, Selangor, Malaysia
| | - Azizah Abdul Hamid
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang, 43300, Selangor, Malaysia
| | - Barakatun-Nisak Mohd Yusof
- Department of Nutrition and Dietetics, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
- Research Centre of Excellent for Nutrition and Non-communicable Diseases, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43300, Selangor, Malaysia.
| |
Collapse
|
34
|
Black PN, Ahowesso C, Montefusco D, Saini N, DiRusso CC. Fatty Acid Transport Proteins: Targeting FATP2 as a Gatekeeper Involved in the Transport of Exogenous Fatty Acids. MEDCHEMCOMM 2016; 7:612-622. [PMID: 27446528 DOI: 10.1039/c6md00043f] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The fatty acid transport proteins (FATP) are classified as members of the Solute Carrier 27 (Slc27) family of proteins based on their ability to function in the transport of exogenous fatty acids. These proteins, when localized to the plasma membrane or at intracellular membrane junctions with the endoplasmic reticulum, function as a gate in the regulated transport of fatty acids and thus represent a therapeutic target to delimit the acquisition of fatty acids that contribute to disease as in the case of fatty acid overload. To date, FATP1, FATP2, and FATP4 have been used as targets in the selection of small molecule inhibitors with the goal of treating insulin resistance and attenuating dietary absorption of fatty acids. Several studies targeting FATP1 and FATP4 were based on the intrinsic acyl CoA synthetase activity of these proteins and not on transport directly. While several classes of compounds were identified as potential inhibitors of fatty acid transport, in vivo studies using a mouse model failed to provide evidence these compounds were effective in blocking or attenuating fatty acid transport. Studies targeting FATP2 employed a naturally occurring splice variant, FATP2b, which lacks intrinsic acyl CoA synthetase due to the deletion of exon 3, yet is fully functional in fatty acid transport. These studies identified two compounds, 5'-bromo-5-phenyl-spiro[3H-1,3,4-thiadiazole-2,3'-indoline]-2'-one), now referred to as Lipofermata, and 2-benzyl-3-(4-chlorophenyl)-5-(4-nitrophenyl)pyrazolo[1,5-a]pyrimidin-7(4H)-one, now called Grassofermata, that are effective fatty acid transport inhibitors both in vitro using a series of model cell lines and in vivo using a mouse model.
Collapse
Affiliation(s)
- Paul N Black
- Department of Biochemistry, University of Nebraska, Lincoln, NE
| | | | | | - Nipun Saini
- Department of Biochemistry, University of Nebraska, Lincoln, NE
| | | |
Collapse
|
35
|
Oh DY, Olefsky JM. G protein-coupled receptors as targets for anti-diabetic therapeutics. Nat Rev Drug Discov 2016; 15:161-72. [PMID: 26822831 DOI: 10.1038/nrd.2015.4] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
36
|
Eight Weeks of Cosmos caudatus (Ulam Raja) Supplementation Improves Glycemic Status in Patients with Type 2 Diabetes: A Randomized Controlled Trial. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:405615. [PMID: 26713097 PMCID: PMC4680046 DOI: 10.1155/2015/405615] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 11/05/2015] [Accepted: 11/08/2015] [Indexed: 12/25/2022]
Abstract
Objectives. Optimizing glycemic control is crucial to prevent type 2 diabetes related complications. Cosmos caudatus is reported to have promising effect in improving plasma blood glucose in an animal model. However, its impact on human remains ambiguous. This study was carried out to evaluate the effectiveness of C. caudatus on glycemic status in patients with type 2 diabetes. Materials and Methods. In this randomized controlled trial with two-arm parallel-group design, a total of 101 subjects with type 2 diabetes were randomly allocated to diabetic-ulam or diabetic controls for eight weeks. Subjects in diabetic-ulam group consumed 15 g of C. caudatus daily for eight weeks while diabetic controls abstained from taking C. caudatus. Both groups received the standard lifestyle advice. Results. After 8 weeks of supplementation, C. caudatus significantly reduced serum insulin (-1.16 versus +3.91), reduced HOMA-IR (-1.09 versus +1.34), and increased QUICKI (+0.05 versus -0.03) in diabetic-ulam group compared with the diabetic controls. HbA1C level was improved although it is not statistically significant (-0.76% versus -0.37%). C. caudatus was safe to consume. Conclusions. C. caudatus supplementation significantly improves insulin resistance and insulin sensitivity in patients with type 2 diabetes.
Collapse
|
37
|
Ali MA, El-Abhar HS, Kamel MA, Attia AS. Antidiabetic Effect of Galantamine: Novel Effect for a Known Centrally Acting Drug. PLoS One 2015; 10:e0134648. [PMID: 26262991 PMCID: PMC4532414 DOI: 10.1371/journal.pone.0134648] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 07/10/2015] [Indexed: 01/26/2023] Open
Abstract
The cholinergic anti-inflammatory pathway is one of the putative biochemical pathways that link diabetes with Alzheimer disease. Hence, we aimed to verify the potential antidiabetic effect of galantamine, unveil the possible mechanisms and evaluate its interaction with vildagliptin. The n5-STZ rat model was adopted and the diabetic animals were treated with galantamine and/or vildagliptin for 4 weeks. Galantamine lowered the n5-STZ-induced elevation in body weight, food/water intake, serum levels of glucose, fructosamine, and ALT/AST, as well as AChE in the tested organs. Moreover, it modulated successfully the lipid profile assessed in serum, liver, and muscle, and increased serum insulin level, as well as % β-cell function, in a pattern similar to that of vildagliptin. Additionally, galantamine confirmed its antioxidant (Nrf2, TAC, MDA), anti-inflammatory (NF-κB, TNF-α, visfatin, adiponectin) and anti-apoptotic (caspase-3, cytochrome c) capabilities by altering the n5-STZ effect on all the aforementioned parameters. On the molecular level, galantamine/vildagliptin have improved the insulin (p-insulin receptor, p-Akt, GLUT4/GLUT2) and Wnt/β-catenin (p-GSK-3β, β-catenin) signaling pathways. On almost all parameters, the galantamine effects surpassed that of vildagliptin, while the combination regimen showed the best effects. The present results clearly proved that galantamine modulated glucose/lipid profile possibly through its anti-oxidant, -apoptotic, -inflammatory and -cholinesterase properties. These effects could be attributed partly to the enhancement of insulin and Wnt/β-catenin signaling pathways. Galantamine can be strongly considered as a potential antidiabetic agent and as an add-on therapy with other oral antidiabetics.
Collapse
Affiliation(s)
- Mennatallah A. Ali
- Department of Pharmacology & Toxicology, Faculty of Pharmacy and Drug Manufacturing, Pharos University, Alexandria, Egypt
| | - Hanan S. El-Abhar
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Maher A. Kamel
- Department of Biochemistry, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Ahmed S. Attia
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
38
|
Tabatabaei-Malazy O, Larijani B, Abdollahi M. Targeting metabolic disorders by natural products. J Diabetes Metab Disord 2015; 14:57. [PMID: 26157708 PMCID: PMC4495701 DOI: 10.1186/s40200-015-0184-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 06/22/2015] [Indexed: 12/15/2022]
Abstract
The most prevalent metabolic disorders are diabetes mellitus, obesity, dyslipidemia, osteoporosis and metabolic syndrome, which are developed when normal metabolic processes are disturbed. The most common pathophysiologies of the above disorders are oxidative stress, Nrf2 pathways, epigenetic, and change in miRNA expression. There is a challenge in the prevention and treatment of metabolic disorders due to severe adverse effects of some synthetic drugs, their high cost, lack of safety and poverty in some conditions, and insufficient accessibility for the general population in the world. With increasing interest in shifting from synthetic drugs to phytotherapy as an alternative treatment, there is still a gap in scientific evidences of plant-derived therapeutic benefits. One reason may be slow rate of translation of animal studies' findings into human clinical trials. Since metabolic disorders are multifactorial, it seems that poly-herbal medications, or drug-herbal combination are needed for their treatment. However, further researches to determine the most effective plant-derived metabolites, and their cellular mechanism in order to set priorities for well-designed animal and clinical trials, and also more studies with strong scientific evidences such as systematic review and meta-analysis of controlled studies are needed.
Collapse
Affiliation(s)
- Ozra Tabatabaei-Malazy
- />Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- />Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- />Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abdollahi
- />Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- />Department of Toxicology and Pharmacology, Faculty of Pharmacy, and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
39
|
Cerf ME. High fat programming of beta cell compensation, exhaustion, death and dysfunction. Pediatr Diabetes 2015; 16:71-8. [PMID: 25682938 DOI: 10.1111/pedi.12137] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 01/07/2014] [Accepted: 02/13/2014] [Indexed: 12/29/2022] Open
Abstract
Programming refers to events during critical developmental windows that shape progeny health outcomes. Fetal programming refers to the effects of intrauterine (in utero) events. Lactational programming refers to the effects of events during suckling (weaning). Developmental programming refers to the effects of events during both fetal and lactational life. Postnatal programming refers to the effects of events either from birth (lactational life) to adolescence or from weaning (end of lactation) to adolescence. Islets are most plastic during the early life course; hence programming during fetal and lactational life is most potent. High fat (HF) programming is the maintenance on a HF diet (HFD) during critical developmental life stages that alters progeny metabolism and physiology. HF programming induces variable diabetogenic phenotypes dependent on the timing and duration of the dietary insult. Maternal obesity reinforces HF programming effects in progeny. HF programming, through acute hyperglycemia, initiates beta cell compensation. However, HF programming eventually leads to chronic hyperglycemia that triggers beta cell exhaustion, death and dysfunction. In HF programming, beta cell dysfunction often co-presents with insulin resistance. Balanced, healthy nutrition during developmental windows is critical for preserving beta cell structure and function. Thus early positive nutritional interventions that coincide with the development of beta cells may reduce the overwhelming burden of diabetes and metabolic disease.
Collapse
Affiliation(s)
- Marlon E Cerf
- Diabetes Discovery Platform, Medical Research Council, Tygerberg, Cape Town, South Africa
| |
Collapse
|
40
|
Effects of berberine on amelioration of hyperglycemia and oxidative stress in high glucose and high fat diet-induced diabetic hamsters in vivo. BIOMED RESEARCH INTERNATIONAL 2015; 2015:313808. [PMID: 25705654 PMCID: PMC4331319 DOI: 10.1155/2015/313808] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 12/20/2014] [Accepted: 12/22/2014] [Indexed: 02/02/2023]
Abstract
This study investigated the effects of berberine on amelioration of hyperglycemia and hyperlipidemia and the mechanism involved in high glucose and high fat diet-induced diabetic hamsters. Golden hamsters fed with high glucose and high fat diet were medicated with metformin, simvastatin, and low or high dose of berberine (50 and 100 mg·kg(-1)) for 6 weeks. The results showed that the body weights were significantly lower in berberine-treated groups than control group. Histological analyses revealed that the treatment of berberine inhibited hepatic fat accumulation. Berberine significantly reduced plasma total cholesterol, triglyceride, free fatty acid, low density lipoprotein cholesterol, malondialdehyde, thiobarbituric acid-reactive substance, and 8-isoprostane level but significantly increased plasma superoxide dismutase activity. Glucose and insulin levels were significantly reduced in metformin and berberine-treated groups. Glucose tolerance tests documented that berberine-treated mice were more glucose tolerant. Berberine treatment increased expression of skeletal muscle glucose transporter 4 mRNA and significantly decreased liver low density lipoprotein receptor mRNA expression. The study suggested that berberine was effective in lowering blood glucose and lipids levels, reducing the body weight, and alleviating the oxidative stress in diabetic hamsters, which might be beneficial in reducing the cardiovascular risk factors in diabetes.
Collapse
|
41
|
Yida Z, Imam MU, Ismail M, Ooi DJ, Sarega N, Azmi NH, Ismail N, Chan KW, Hou Z, Yusuf NB. Edible Bird's Nest Prevents High Fat Diet-Induced Insulin Resistance in Rats. J Diabetes Res 2015; 2015:760535. [PMID: 26273674 PMCID: PMC4530265 DOI: 10.1155/2015/760535] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 03/10/2015] [Accepted: 03/17/2015] [Indexed: 01/22/2023] Open
Abstract
Edible bird's nest (EBN) is used traditionally in many parts of Asia to improve wellbeing, but there are limited studies on its efficacy. We explored the potential use of EBN for prevention of high fat diet- (HFD-) induced insulin resistance in rats. HFD was given to rats with or without simvastatin or EBN for 12 weeks. During the intervention period, weight measurements were recorded weekly. Blood samples were collected at the end of the intervention and oral glucose tolerance test conducted, after which the rats were sacrificed and their liver and adipose tissues collected for further studies. Serum adiponectin, leptin, F2-isoprostane, insulin, and lipid profile were estimated, and homeostatic model assessment of insulin resistance computed. Effects of the different interventions on transcriptional regulation of insulin signaling genes were also evaluated. The results showed that HFD worsened metabolic indices and induced insulin resistance partly through transcriptional regulation of the insulin signaling genes. Additionally, simvastatin was able to prevent hypercholesterolemia but promoted insulin resistance similar to HFD. EBN, on the other hand, prevented the worsening of metabolic indices and transcriptional changes in insulin signaling genes due to HFD. The results suggest that EBN may be used as functional food to prevent insulin resistance.
Collapse
Affiliation(s)
- Zhang Yida
- Laboratory of Molecular Biomedicine, Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
- Cardiology Department, Affiliated Hospital of Chengde Medical University, Chengde, Hebei 067000, China
| | - Mustapha Umar Imam
- Laboratory of Molecular Biomedicine, Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Maznah Ismail
- Laboratory of Molecular Biomedicine, Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
- Department of Nutrition and Dietetics, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
- *Maznah Ismail:
| | - Der-Jiun Ooi
- Laboratory of Molecular Biomedicine, Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Nadarajan Sarega
- Laboratory of Molecular Biomedicine, Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Nur Hanisah Azmi
- Laboratory of Molecular Biomedicine, Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Norsharina Ismail
- Laboratory of Molecular Biomedicine, Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Kim Wei Chan
- Laboratory of Molecular Biomedicine, Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Zhiping Hou
- Laboratory of Molecular Biomedicine, Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Norhayati Binti Yusuf
- Laboratory of Molecular Biomedicine, Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| |
Collapse
|
42
|
VandenBerg P. The Canadian Diabetes Association, Canadian Society, is funding CDN $7.5 million in research in 2014-2015 to support excellent researchers and research trainees. Can J Diabetes 2014; 38:393-5. [PMID: 25449553 DOI: 10.1016/j.jcjd.2014.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 09/17/2014] [Indexed: 11/30/2022]
Affiliation(s)
- Polly VandenBerg
- Manager, Research Knowledge Translation, Canadian Diabetes Association, Toronto, Ontario, Canada
| |
Collapse
|
43
|
Li G, Kong L, Zhang L, Fan L, Su Y, Rose JC, Zhang W. Early Pregnancy Maternal Lipid Profiles and the Risk of Gestational Diabetes Mellitus Stratified for Body Mass Index. Reprod Sci 2014; 22:712-7. [PMID: 25394643 DOI: 10.1177/1933719114557896] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To determine associations between lipid profiles in early pregnancy stratified by body mass index (BMI) and risk of developing gestational diabetes mellitus (GDM). STUDY DESIGN A total of 2488 healthy pregnant women were enrolled prospectively. Fasting plasma lipid profiles were measured at mean 11 weeks of gestation including triglycerides (TGs), total cholesterol, high-density lipoprotein (HDL), low-density lipoprotein (LDL), and cholesterol (CHO). We assessed early pregnancy maternal lipid concentrations in different tertiles in association with the risk of GDM stratified for BMI. Multivariable logistic regression analyses were used to estimate the relative risk of GDM by calculating odds ratios and 95% confidence intervals (CIs). RESULTS In univariate analyses, pregnant women with GDM had significantly increased serum TG, CHO, LDL concentrations, LDL/HDL ratio, and decreased LDL concentrations, compared to control groups, each P < .01, respectively. After adjustment for confounders, there was a 1.8-fold increase in risk for GDM in the lean group (95% CI: 1.2-2.7) and 2.7-fold increase in the obese group (95% CI: 1.1-6.6), respectively, if TG ≥ 1.58 mmol/L. About a 50% decrease in the risk of GDM was observed in lean women with HDL ≥ 2.22 mmol/L (95% CI: 0.3-0.9). No significant correlations of other lipid profiles with the risk of developing GDM were observed. CONCLUSION Early pregnancy dyslipidemia is associated with the risk of developing GDM. Lean or obese women with higher TG concentrations are at an increased risk for developing GDM while lean women with high HDL are protected.
Collapse
Affiliation(s)
- Guanghui Li
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Lijun Kong
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Li Zhang
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Ling Fan
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Yixin Su
- Department of Obstetrics and Gynecology, Wake Forest University School of Medicine, NC, USA
| | - James C Rose
- Department of Obstetrics and Gynecology, Wake Forest University School of Medicine, NC, USA
| | - Weiyuan Zhang
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
44
|
Agrawal NK, Kant S. Targeting inflammation in diabetes: Newer therapeutic options. World J Diabetes 2014; 5:697-710. [PMID: 25317247 PMCID: PMC4138593 DOI: 10.4239/wjd.v5.i5.697] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 04/24/2014] [Accepted: 05/29/2014] [Indexed: 02/05/2023] Open
Abstract
Inflammation has been recognised to both decrease beta cell insulin secretion and increase insulin resistance. Circulating cytokines can affect beta cell function directly leading to secretory dysfunction and increased apoptosis. These cytokines can also indirectly affect beta cell function by increasing adipocyte inflammation.The resulting glucotoxicity and lipotoxicity further enhance the inflammatory process resulting in a vicious cycle. Weight reduction and drugs such as metformin have been shown to decrease the levels of C-Reactive Protein by 31% and 13%, respectively. Pioglitazone, insulin and statins have anti-inflammatory effects. Interleukin 1 and tumor necrosis factor-α antagonists are in trials and NSAIDs such as salsalate have shown an improvement in insulin sensitivity. Inhibition of 12-lipo-oxygenase, histone de-acetylases, and activation of sirtuin-1 are upcoming molecular targets to reduce inflammation. These therapies have also been shown to decrease the conversion of pre-diabetes state to diabetes. Drugs like glicazide, troglitazone, N-acetylcysteine and selective COX-2 inhibitors have shown benefit in diabetic neuropathy by decreasing inflammatory markers. Retinopathy drugs are used to target vascular endothelial growth factor, angiopoietin-2, various proteinases and chemokines. Drugs targeting the proteinases and various chemokines are pentoxifylline, inhibitors of nuclear factor-kappa B and mammalian target of rapamycin and are in clinical trials for diabetic nephropathy. Commonly used drugs such as insulin, metformin, peroxisome proliferator-activated receptors, glucagon like peptide-1 agonists and dipeptidyl peptidase-4 inhibitors also decrease inflammation. Anti-inflammatory therapies represent a potential approach for the therapy of diabetes and its complications.
Collapse
|
45
|
Hornbak M, Allin KH, Jensen ML, Lau CJ, Witte D, Jørgensen ME, Sandbæk A, Lauritzen T, Andersson Å, Pedersen O, Hansen T. A combined analysis of 48 type 2 diabetes genetic risk variants shows no discriminative value to predict time to first prescription of a glucose lowering drug in Danish patients with screen detected type 2 diabetes. PLoS One 2014; 9:e104837. [PMID: 25157406 PMCID: PMC4144838 DOI: 10.1371/journal.pone.0104837] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 07/03/2014] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE To investigate the genetic influence of 48 type 2 diabetes susceptibility variants on disease progression measured as risk of early prescription redemption of glucose lowering drugs in screen-detected patients with type 2 diabetes. METHODS We studied type 2 diabetes progression in 1,480 patients with screen-detected type 2 diabetes from the ADDITION-Denmark study using information of redeemed prescriptions from the Register of Medicinal Products Statistics from 2001-2009 in Denmark. Patients were cluster randomized by general practitioners, who were randomized to treat type 2 diabetes according to either a conventional or a multifactorial intensive treatment algorithm. We investigated the genetic influence on diabetes progression by constructing a genetic risk score (GRS) of all 48 validated type 2 diabetes susceptibility variants, a GRS of 11 variants linked to β-cell function and a GRS of 3 variants linked to insulin sensitivity and assessed the association between number of risk alleles and time from diagnosis until first redeemed prescription of either any glucose lowering drug or an insulin drug. RESULTS The GRS linked to insulin sensitivity only nominally increased the risk of an early prescription redemption with an insulin drug by 39% (HR [95% C.I.] = 1.39 [1.09-1.77], p = 0.009] in patients randomized to the intensive treatment group. Furthermore, the strongest univariate predictors of diabetes progression for the intensive treatment group (measured as time to first insulin) were younger age (HR [95% C.I.] = 0.96 [0.93-0.99]), increased BMI (1.05 [1.01-1.09]), increased HbA1c (1.50 [1.36-.66]), increased TG (1.24 [1.11-1.39]) and reduced fasting serum HDL (0.37 [0.17-0.80]) at baseline. Similar results were obtained for the conventional treatment group. CONCLUSION Higher levels of HbA1c, fasting circulating levels of triglyceride, lower HDL, larger BMI and younger age are significant determinants of early pharmacological intervention in type 2 diabetes. However, known common type 2 diabetes-associated gene variants do not appear to significantly affect disease progression.
Collapse
Affiliation(s)
- Malene Hornbak
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- School of Pharmaceutical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| | - Kristine Højgaard Allin
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Majken Linnemann Jensen
- Steno Diabetes Center A/S, Gentofte, Denmark
- Section for Social and Clinical Pharmacy, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Cathrine Juel Lau
- Research Centre for Prevention and Health, Capital Region of Denmark, Glostrup Hospital, Glostrup, Denmark
| | - Daniel Witte
- Public Research Centre for Health, Centre for Health Studies, Strassen, Luxembourg
| | | | - Annelli Sandbæk
- Department of Public Health, Section of General Practice Medicine, Aarhus University, Aarhus, Denmark
| | - Torsten Lauritzen
- Department of Public Health, Section of General Practice Medicine, Aarhus University, Aarhus, Denmark
| | - Åsa Andersson
- School of Pharmaceutical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Oluf Pedersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Institute of Biomedical Science, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Faculty of Health Sciences, University of Aarhus, Aarhus, Denmark
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Section of Molecular Diabetes & Metabolism, Institute of Clinical Research & Institute of Molecular Medicine, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
46
|
Cernea S, Dobreanu M. Diabetes and beta cell function: from mechanisms to evaluation and clinical implications. Biochem Med (Zagreb) 2013; 23:266-80. [PMID: 24266296 PMCID: PMC3900074 DOI: 10.11613/bm.2013.033] [Citation(s) in RCA: 133] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Diabetes is a complex, heterogeneous condition that has beta cell dysfunction at its core. Many factors (e.g. hyperglycemia/glucotoxicity, lipotoxicity, autoimmunity, inflammation, adipokines, islet amyloid, incretins and insulin resistance) influence the function of pancreatic beta cells. Chronic hyperglycaemia may result in detrimental effects on insulin synthesis/secretion, cell survival and insulin sensitivity through multiple mechanisms: gradual loss of insulin gene expression and other beta-cell specific genes; chronic endoplasmic reticulum stress and oxidative stress; changes in mitochondrial number, morphology and function; disruption in calcium homeostasis. In the presence of hyperglycaemia, prolonged exposure to increased free fatty acids result in accumulation of toxic metabolites in the cells (“lipotoxicity”), finally causing decreased insulin gene expression and impairment of insulin secretion. The rest of the factors/mechanisms which impact on the course of the disease are also discusses in detail. The correct assessment of beta cell function requires a concomitant quantification of insulin secretion and insulin sensitivity, because the two variables are closely interrelated. In order to better understand the fundamental pathogenetic mechanisms that contribute to disease development in a certain individual with diabetes, additional markers could be used, apart from those that evaluate beta cell function. The aim of the paper was to overview the relevant mechanisms/factors that influence beta cell function and to discuss the available methods of its assessment. In addition, clinical considerations are made regarding the therapeutical options that have potential protective effects on beta cell function/mass by targeting various underlying factors and mechanisms with a role in disease progression.
Collapse
Affiliation(s)
- Simona Cernea
- Diabetes, Nutrition and Metabolic Diseases Outpatient Unit, Emergency County Clinical Hospital, Târgu Mureş, Romania.
| | | |
Collapse
|
47
|
Wu J, Wu JJ, Yang LJ, Wei LX, Zou DJ. Rosiglitazone protects against palmitate-induced pancreatic beta-cell death by activation of autophagy via 5'-AMP-activated protein kinase modulation. Endocrine 2013; 44:87-98. [PMID: 23109223 DOI: 10.1007/s12020-012-9826-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 10/19/2012] [Indexed: 01/20/2023]
Abstract
Promoting beta-cell survival is crucial for the prevention of beta-cell failure in diabetes. Thiazolidinediones, a widely used drug to improve insulin sensitivity in clinical practice, is found to have a protective effect on islet beta-cell. To date, the mechanism underlying the protective role of thiazolidinedione on beta-cell survival remain largely unknown. Activation of autophagy was detected by transmission electron microscopy, western blot, and GFP-LC3 transfection. Cell viability was examined by WST-8. Cell apoptosis was demonstrated by DAPI and Annexin V/PI staining. Colony formation assay was used to detect long-term cell viability. We demonstrated that rosiglitazone-treated beta-cells were more resistant to palmitate-induced apoptosis. The conversion of LC3-I to LC3-II and accumulated autophagosomes were found to be upregulated in rosiglitazone-treated cells. Inhibition of autophagy augmented palmitate-induced apoptosis with rosiglitazone treatment, suggesting that autophagy plays an important role in the survival function of rosiglitazone on beta-cells. Furthermore, we showed that rosiglitazone could induce AMP-activated protein kinase (AMPK) phosphorylation and reduce p70S6 kinase phosphorylation. Inhibition of AMPK impaired autophagy activation and enhanced palmitate-induced apoptosis during rosiglitazone treatment. These findings reveal that rosiglitazone-induced autophagy contributes to its protective function on beta-cells during palmitate treatment.
Collapse
Affiliation(s)
- Jie Wu
- Department of Endocrinology, Chang Hai Hospital, The Second Military Medical University, 168 Changhai Road, Shanghai, 200438, People's Republic of China
| | | | | | | | | |
Collapse
|
48
|
Akash MSH, Rehman K, Chen S. Role of inflammatory mechanisms in pathogenesis of type 2 diabetes mellitus. J Cell Biochem 2013; 114:525-31. [PMID: 22991242 DOI: 10.1002/jcb.24402] [Citation(s) in RCA: 255] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 09/11/2012] [Indexed: 12/12/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is characterized by progressive β-cell dysfunctioning and insulin resistance. This article reviews recent literature with special focus on inflammatory mechanisms that provoke the pathogenesis of T2DM. We have focused on the recent advances in progression of T2DM including various inflammatory mechanisms that might induce inflammation, insulin resistance, decrease insulin secretion from pancreatic islets and dysfunctioning of β-cells. Here we have also summarized the role of various pro-inflammatory mediators involved in inflammatory mechanisms, which may further alter the normal structure of β-cells by inducing pancreatic islet's apoptosis. In conclusion, it is suggested that the role of inflammation in pathogenesis of T2DM is crucial and cannot be neglected. Moreover, the insight of inflammatory responses in T2DM may provide a new gateway for the better treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Muhammad Sajid Hamid Akash
- Institute of Pharmacology, Toxicology, and Biochemical Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | | | | |
Collapse
|
49
|
Imai Y, Dobrian AD, Morris MA, Nadler JL. Islet inflammation: a unifying target for diabetes treatment? Trends Endocrinol Metab 2013; 24:351-60. [PMID: 23484621 PMCID: PMC3686848 DOI: 10.1016/j.tem.2013.01.007] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 01/22/2013] [Accepted: 01/23/2013] [Indexed: 12/12/2022]
Abstract
In the past decade, islet inflammation has emerged as a contributor to the loss of functional β cell mass in both type 1 (T1D) and type 2 diabetes (T2D). Evidence supports the idea that overnutrition and insulin resistance result in the production of proinflammatory mediators by β cells. In addition to compromising β cell function and survival, cytokines may recruit macrophages into islets, thus augmenting inflammation. Limited but intriguing data imply a role of adaptive immune response in islet dysfunction in T2D. Clinical trials have validated anti-inflammatory therapies in T2D, whereas immune therapy for T1D remains challenging. Further research is required to improve our understanding of islet inflammatory pathways and to identify more effective therapeutic targets for T1D and T2D.
Collapse
Affiliation(s)
- Yumi Imai
- Department of Internal Medicine, Strelitz Diabetes Center, Eastern Virginia Medical School, Norfolk, VA 23507, USA.
| | | | | | | |
Collapse
|
50
|
Spanou M, Tziomalos K. Bariatric surgery as a treatment option in patients with type 2 diabetes mellitus. World J Diabetes 2013; 4:14-18. [PMID: 23593531 PMCID: PMC3627414 DOI: 10.4239/wjd.v4.i2.14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Revised: 02/10/2013] [Accepted: 03/06/2013] [Indexed: 02/05/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a leading cause of blindness, non-traumatic amputation and end-stage renal disease as well as a major cardiovascular risk factor. Tight glycemic control reduces the incidence of microvascular complications of T2DM whereas its effects on macrovascular complication are more controversial. However, glycemic targets are achieved by a minority of diabetic patients despite the availability of several antidiabetic agents. In the present commentary, we discuss the findings of two recent randomized studies that compared bariatric surgery with medical treatment in patients with uncontrolled T2DM. Both studies showed that bariatric surgery results in remission of T2DM in the majority of patients. However, both studies were limited to relatively young patients without comorbidities, had relatively short follow-up and did not assess the effects of surgery on T2DM complications. Moreover, the perioperative complications of bariatric surgery and its limited availability in some areas are additional barriers to the wider implementation of this therapeutic approach. On the other hand, the elucidation of the mechanisms underpinning the resolution of T2DM following bariatric surgery might result in the development of novel, more effective pharmacotherapies for this common disease.
Collapse
|