1
|
Sokolowski EK, Kursawe R, Selvam V, Bhuiyan RM, Thibodeau A, Zhao C, Spracklen CN, Ucar D, Stitzel ML. Multi-omic human pancreatic islet endoplasmic reticulum and cytokine stress response mapping provides type 2 diabetes genetic insights. Cell Metab 2024; 36:2468-2488.e7. [PMID: 39383866 DOI: 10.1016/j.cmet.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 06/14/2024] [Accepted: 09/10/2024] [Indexed: 10/11/2024]
Abstract
Endoplasmic reticulum (ER) and inflammatory stress responses contribute to islet dysfunction in type 2 diabetes (T2D). Comprehensive genomic understanding of these human islet stress responses and whether T2D-associated genetic variants modulate them is lacking. Here, comparative transcriptome and epigenome analyses of human islets exposed ex vivo to these stressors revealed 30% of expressed genes and 14% of islet cis-regulatory elements (CREs) as stress responsive, modulated largely in an ER- or cytokine-specific fashion. T2D variants overlapped 86 stress-responsive CREs, including 21 induced by ER stress. We linked the rs6917676-T T2D risk allele to increased islet ER-stress-responsive CRE accessibility and allele-specific β cell nuclear factor binding. MAP3K5, the ER-stress-responsive putative rs6917676 T2D effector gene, promoted stress-induced β cell apoptosis. Supporting its pro-diabetogenic role, MAP3K5 expression correlated inversely with human islet β cell abundance and was elevated in T2D β cells. This study provides genome-wide insights into human islet stress responses and context-specific T2D variant effects.
Collapse
Affiliation(s)
- Eishani K Sokolowski
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Romy Kursawe
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Vijay Selvam
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Redwan M Bhuiyan
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Asa Thibodeau
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Chi Zhao
- Department of Biostatistics and Epidemiology, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Cassandra N Spracklen
- Department of Biostatistics and Epidemiology, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA; Institute of Systems Genomics, University of Connecticut, Farmington, CT 06032, USA.
| | - Michael L Stitzel
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA; Institute of Systems Genomics, University of Connecticut, Farmington, CT 06032, USA.
| |
Collapse
|
2
|
Velagapudi S, Karsai G, Karsai M, Mohammed SA, Montecucco F, Liberale L, Lee H, Carbone F, Adami GF, Yang K, Crucet M, Stein S, Paneni F, Lapikova-Bryhinska T, Jang HD, Kraler S, Vdovenko D, Züllig RA, Camici GG, Kim HS, Laaksonen R, Gerber PA, Hornemann T, Akhmedov A, Lüscher TF. Inhibition of de novo ceramide synthesis by sirtuin-1 improves beta-cell function and glucose metabolism in type 2 diabetes. Cardiovasc Res 2024; 120:1265-1278. [PMID: 38739545 DOI: 10.1093/cvr/cvae100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 03/03/2024] [Accepted: 03/26/2024] [Indexed: 05/16/2024] Open
Abstract
AIMS Obesity and type 2 diabetes (T2D) are major risk factors for cardiovascular (CV) diseases. Dysregulated pro-apoptotic ceramide synthesis reduces β-cell insulin secretion, thereby promoting hyperglycaemic states that may manifest as T2D. Pro-apoptotic ceramides modulate insulin sensitivity and glucose tolerance while being linked to poor CV outcomes. Sirtuin-1 (SIRT1) is a NAD + -dependent deacetylase that protects against pancreatic β-cell dysfunction; however, systemic levels are decreased in obese-T2D mice and may promote pro-apoptotic ceramide synthesis and hyperglycaemia. Herein, we aimed to assess the effects of restoring circulating SIRT1 levels to prevent metabolic imbalance in obese and diabetic mice. METHODS AND RESULTS Circulating SIRT1 levels were reduced in obese-diabetic mice (db/db) as compared to age-matched non-diabetic db/+ controls. Restoration of SIRT1 plasma levels with recombinant murine SIRT1 for 4 weeks prevented body weight gain and improved glucose tolerance, insulin sensitivity, and vascular function in mice models of obesity and T2D. Untargeted lipidomics revealed that SIRT1 restored insulin secretory function of β-cells by reducing synthesis and accumulation of pro-apoptotic ceramides. Molecular mechanisms involved direct binding to and deacetylation of Toll-like receptor 4 (TLR4) by SIRT1 in β-cells, thereby decreasing the rate-limiting enzymes of sphingolipid synthesis SPTLC1/2 via AKT/NF-κB. Among patients with T2D, those with high baseline plasma levels of SIRT1 prior to metabolic surgery displayed restored β-cell function (HOMA2-β) and were more likely to have T2D remission during follow-up. CONCLUSION Acetylation of TLR4 promotes β-cell dysfunction via ceramide synthesis in T2D, which is blunted by systemic SIRT1 replenishment. Hence, restoration of systemic SIRT1 may provide a novel therapeutic strategy to counteract toxic ceramide synthesis and mitigate CV complications of T2D.
Collapse
Affiliation(s)
- Srividya Velagapudi
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
| | - Gergely Karsai
- Institute of Clinical Chemistry, University Hospital Zürich, Zürich, Switzerland
| | - Maria Karsai
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zürich and University of Zürich, Zürich, Switzerland
| | - Shafeeq A Mohammed
- Department of Cardiology, Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital and University of Zürich, Zürich, Switzerland
| | - Fabrizio Montecucco
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa School of Medicine, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Genoa, Italy
| | - Luca Liberale
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa School of Medicine, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Genoa, Italy
| | - Hwan Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Federico Carbone
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa School of Medicine, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Genoa, Italy
| | - Giovanni Francesco Adami
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa School of Medicine, Genoa, Italy
| | - Kangmin Yang
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
| | - Margot Crucet
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
| | - Sokrates Stein
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
| | - Franceso Paneni
- Department of Cardiology, Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital and University of Zürich, Zürich, Switzerland
| | | | - Hyun-Duk Jang
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
| | - Daria Vdovenko
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
| | - Richard Arnold Züllig
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zürich and University of Zürich, Zürich, Switzerland
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
| | - Hyo-Soo Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Reijo Laaksonen
- Zora Biosciences and Finnish Cardiovascular Research Center, Finland Medical School, Tampere University, Tampere, Finland
| | - Philipp A Gerber
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zürich and University of Zürich, Zürich, Switzerland
| | - Thorsten Hornemann
- Institute of Clinical Chemistry, University Hospital Zürich, Zürich, Switzerland
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
- Royal Brompton and Harefield Hospitals, Imperial College and King's College, London, United Kingdom
| |
Collapse
|
3
|
Johnson-Pitt A, Catchpole B, Davison LJ. Exocrine pancreatic inflammation in canine diabetes mellitus - An active offender? Vet J 2024; 308:106241. [PMID: 39243807 DOI: 10.1016/j.tvjl.2024.106241] [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: 06/05/2024] [Revised: 08/27/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024]
Abstract
The purpose of this review is to examine the current scientific literature regarding the interplay between the exocrine and endocrine pancreas, specifically the role of the exocrine pancreas in the pathogenesis of canine diabetes mellitus. β-cell death caused by exocrine pancreatic inflammation is thought to be an under-recognised contributor to diabetes mellitus in dogs, with up to 30 % of canine diabetic patients with concurrent evidence of pancreatitis at post-mortem examination. Current diagnostics for pancreatitis are imprecise, and treatments for both diseases individually have their own limitations: diabetes through daily insulin injections, which has both welfare and financial implications for the stakeholders, and pancreatitis through treatment of clinical signs, such as analgesia and anti-emetics, rather than targeted treatment of the underlying cause. This review will consider the evidence for exocrine pancreatic inflammation making an active contribution to pancreatic β-cell loss and insulin-deficiency diabetes in the dog and explore current and potential future diagnostic and treatment avenues to improve outcomes for these patients.
Collapse
Affiliation(s)
- Arielle Johnson-Pitt
- Department of Clinical Science and Services, The Royal Veterinary College, Hertfordshire AL9 7TA, UK.
| | - Brian Catchpole
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hertfordshire AL9 7TA, UK
| | - Lucy J Davison
- Department of Clinical Science and Services, The Royal Veterinary College, Hertfordshire AL9 7TA, UK; Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| |
Collapse
|
4
|
Han HW, Pradhan G, Villarreal D, Kim DM, Jain A, Gaharwar A, Tian Y, Guo S, Sun Y. GHSR Deletion in β-Cells of Male Mice: Ineffective in Obesity, but Effective in Protecting against Streptozotocin-Induced β-Cell Injury in Aging. Nutrients 2024; 16:1464. [PMID: 38794702 PMCID: PMC11123813 DOI: 10.3390/nu16101464] [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: 04/15/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Insulin secretion from pancreatic β cells is a key pillar of glucose homeostasis, which is impaired under obesity and aging. Growth hormone secretagogue receptor (GHSR) is the receptor of nutrient-sensing hormone ghrelin. Previously, we showed that β-cell GHSR regulated glucose-stimulated insulin secretion (GSIS) in young mice. In the current study, we further investigated the effects of GHSR on insulin secretion in male mice under diet-induced obesity (DIO) and streptozotocin (STZ)-induced β-cell injury in aging. β-cell-specific-Ghsr-deficient (Ghsr-βKO) mice exhibited no glycemic phenotype under DIO but showed significantly improved ex vivo GSIS in aging. We also detected reduced insulin sensitivity and impaired insulin secretion during aging both in vivo and ex vivo. Accordingly, there were age-related alterations in expression of glucose transporter, insulin signaling pathway, and inflammatory genes. To further determine whether GHSR deficiency affected β-cell susceptibility to acute injury, young, middle-aged, and old Ghsr-βKO mice were subjected to STZ. We found that middle-aged and old Ghsr-βKO mice were protected from STZ-induced hyperglycemia and impaired insulin secretion, correlated with increased expression of insulin signaling regulators but decreased pro-inflammatory cytokines in pancreatic islets. Collectively, our findings indicate that β-cell GHSR has a major impact on insulin secretion in aging but not obesity, and GHSR deficiency protects against STZ-induced β-cell injury in aging.
Collapse
Affiliation(s)
- Hye Won Han
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; (H.W.H.)
| | - Geetali Pradhan
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Daniel Villarreal
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; (H.W.H.)
| | - Da Mi Kim
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; (H.W.H.)
| | - Abhishek Jain
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Akhilesh Gaharwar
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Yanan Tian
- Department of Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Shaodong Guo
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; (H.W.H.)
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; (H.W.H.)
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
5
|
Sileikaite-Morvaközi I, Hansen WH, Davies MJ, Mandrup-Poulsen T, Hawkins CL. Detrimental Actions of Chlorinated Nucleosides on the Function and Viability of Insulin-Producing Cells. Int J Mol Sci 2023; 24:14585. [PMID: 37834034 PMCID: PMC10572493 DOI: 10.3390/ijms241914585] [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: 07/26/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Neutrophils are innate immune cells that play a key role in pathogen clearance. They contribute to inflammatory diseases, including diabetes, by releasing pro-inflammatory cytokines, reactive oxygen species, and extracellular traps (NETs). NETs contain a DNA backbone and catalytically active myeloperoxidase (MPO), which produces hypochlorous acid (HOCl). Chlorination of the DNA nucleoside 8-chloro-deoxyguanosine has been reported as an early marker of inflammation in diabetes. In this study, we examined the reactivity of different chlorinated nucleosides, including 5-chloro-(deoxy)cytidine (5ClC, 5CldC), 8-chloro-(deoxy)adenosine (8ClA, 8CldA) and 8-chloro-(deoxy)guanosine (8ClG, 8CldG), with the INS-1E β-cell line. Exposure of INS-1E cells to 5CldC, 8CldA, 8ClA, and 8CldG decreased metabolic activity and intracellular ATP, and, together with 8ClG, induced apoptotic cell death. Exposure to 8ClA, but not the other nucleosides, resulted in sustained endoplasmic reticulum stress, activation of the unfolded protein response, and increased expression of thioredoxin-interacting protein (TXNIP) and heme oxygenase 1 (HO-1). Exposure of INS-1E cells to 5CldC also increased TXNIP and NAD(P)H dehydrogenase quinone 1 (NQO1) expression. In addition, a significant increase in the mRNA expression of NQO1 and GPx4 was seen in INS-1E cells exposed to 8ClG and 8CldA, respectively. However, a significant decrease in intracellular thiols was only observed in INS-1E cells exposed to 8ClG and 8CldG. Finally, a significant decrease in the insulin stimulation index was observed in experiments with all the chlorinated nucleosides, except for 8ClA and 8ClG. Together, these results suggest that increased formation of chlorinated nucleosides during inflammation in diabetes could influence β-cell function and may contribute to disease progression.
Collapse
Affiliation(s)
| | | | | | | | - Clare L. Hawkins
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (I.S.-M.); (M.J.D.); (T.M.-P.)
| |
Collapse
|
6
|
Corkey BE. Reactive oxygen species: role in obesity and mitochondrial energy efficiency. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220210. [PMID: 37482778 PMCID: PMC10363708 DOI: 10.1098/rstb.2022.0210] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 05/29/2023] [Indexed: 07/25/2023] Open
Abstract
Changes correlating with increasing obesity include insulin resistance, hyperlipidaemia, hyperinsulinaemia, highly processed food and environmental toxins including plastics and air pollution. The relationship between the appearance of each of these potential causes and the onset of obesity is unknown. The cause(s) must precede obesity, the consequence, and temporally relate to its rising incidence. Macronutrients such as carbohydrates or fats are unlikely to cause obesity since these have long been constituents of human diets. Furthermore, food consumption and body weight have been well-regulated in most humans and other species until recent times. Thus, attention must focus on changes that have occurred in the last half-century and the relationship between such changes and specific populations that are impacted. The hypothesis presented here is that substances that have entered our bodies recently cause obesity by generating false and misleading information about energy status. We propose that this misinformation is caused by changes in the oxidation-reduction (redox) potential of metabolites that circulate and communicate to organs throughout the body. Examples are provided of food additives that generate reactive oxygen species and impact redox state, thereby, eliciting inappropriate tissue-specific functional changes, including insulin secretion. Reversal requires identification, neutralization, or removal of these compounds. This article is part of a discussion meeting issue 'Causes of obesity: theories, conjectures and evidence (Part I)'.
Collapse
Affiliation(s)
- Barbara E. Corkey
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
7
|
Chang SY, Ko Y, Kim MJ. Regulatory mechanisms of kaempferol on iNOS expression in RINm5F β-cells under exposure to interleukin-1β. Heliyon 2023; 9:e14818. [PMID: 37025778 PMCID: PMC10070653 DOI: 10.1016/j.heliyon.2023.e14818] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 03/17/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
Proinflammatory cytokines and NO play crucial roles in islet β-cells dysfunction. Though anti-inflammatory effects of kaempferol were revealed in several studies, the detailed mechanisms remain unclear. This study explored protective actions of kaempferol in interleukin-1β-treated RINm5F β-cells. Kaempferol significantly inhibited NO generation, iNOS protein, and iNOS mRNA level. Promoter study, EMSA, and κB-dependent reporter assay showed that kaempferol inhibited NF-κB-mediated iNOS gene transcription. Also, we found that kaempferol accelerated iNOS mRNA instability in iNOS 3'-UTR construct and actinomycin D chase studies. Additionally, kaempferol reduced iNOS protein stability in cycloheximide chase study and it inhibited NOS enzyme activity. Kaempferol inhibited ROS generation and preserved cell viability, and it improved insulin release. These findings suggest that kaempferol appears to be helpful in protecting islet β-cells, thereby supports kaempferol as a supplementary therapeutic candidate in inhibiting the incidence and progression of diabetes mellitus.
Collapse
|
8
|
Huang H, Drici L, Lassen PS, Palmisano G, Larsen MR. TiCPG - a strategy for the simultaneous enrichment of reversibly modified cysteine peptides, phosphopeptides, and sialylated N-Glycopeptides to study cytokines stimulated beta-cells. J Proteomics 2023; 273:104796. [PMID: 36538968 DOI: 10.1016/j.jprot.2022.104796] [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: 07/01/2022] [Revised: 12/02/2022] [Accepted: 12/03/2022] [Indexed: 12/23/2022]
Abstract
Diverse post-translational modifications (PTMs) regulate protein function and interaction to fine-tune biological processes. Reversible phosphorylation, cysteines (Cys) modifications, and N-linked glycosylation are all essentially involved in cellular signaling pathways, such as those initiated by the action of pro-inflammatory cytokines, which can induce pancreatic β-cell death and diabetes. Here we have developed a novel strategy for the simultaneous and comprehensive characterization of the proteome and three PTMs including reversibly modified Cysteines (rmCys), phosphorylation, and sialylated N-linked glycosylation from low amount of sample material. This strategy, termed TiCPG, is based on a combination of chemical labeling and titanium dioxide (TiO2) chromatography. We applied the TiCPG strategy to study the proteome and the three PTMs changes in β-cells subject to pro-inflammatory cytokines stimulation. It enabled quantitative analysis of 8346 rmCys sites, 10,321 phosphosites and 962 sialylated N-glycosites from 5496 proteins. Significant regulation was found on 100 proteins at the expression level, while 3020 PTM peptide isoforms from 1468 proteins were significantly regulated. The three PTMs were involved in cytokine mediated β-cell apoptosis, such as the NFκB and the inducible NO synthase signaling pathways. Overall, the TiCPG strategy is a cheap, straightforward, and powerful tool for studies targeting the three PTMs described above. SIGNIFICANCE: The present study presents a fast and easy method for quantitative assessment of the proteome and three PTMs from minimal amount of sample material. This simple method provides comprehensive and significant knowledge on biological systems and cellular signaling with relatively low analysis time, suitable for younger researchers and researchers that do not have direct access to LC-MSMS in their laboratories. From sub-milligram amount of material, we were able to map known cellular signaling events of proinflammatory cytokine effect on beta-cells and to discover novel PTMs involved in several known signaling pathways.
Collapse
Affiliation(s)
- Honggang Huang
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark.
| | - Lylia Drici
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Pernille S Lassen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Giuseppe Palmisano
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark; Departament of Parasitology, Institute of Biomedical Sciences - University of São Paulo, Avenida Prof. Lineu Prestes, 1374 - Edifício Biomédicas II, Cidade Universitária "Armando Salles Oliveira" - CEP, 05508-000 São Paulo, Brazil
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark.
| |
Collapse
|
9
|
Li D, Zhong J, Zhang Q, Zhang J. Effects of anti-inflammatory therapies on glycemic control in type 2 diabetes mellitus. Front Immunol 2023; 14:1125116. [PMID: 36936906 PMCID: PMC10014557 DOI: 10.3389/fimmu.2023.1125116] [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: 12/15/2022] [Accepted: 02/15/2023] [Indexed: 03/05/2023] Open
Abstract
Background The overall evidence base of anti-inflammatory therapies in patients with type 2 diabetes mellitus (T2DM) has not been systematically evaluated. The purpose of this study was to assess the effects of anti-inflammatory therapies on glycemic control in patients with T2DM. Methods PubMed, Embase, Web of Science, and Cochrane Library were searched up to 21 September 2022 for randomized controlled trials (RCTs) with anti-inflammatory therapies targeting the proinflammatory cytokines, cytokine receptors, and inflammation-associated nuclear transcription factors in the pathogenic processes of diabetes, such as interleukin-1β (IL-1β), interleukin-1β receptor (IL-1βR), tumor necrosis factor-α (TNF-α), and nuclear factor-κB (NF-κB). We synthesized data using mean difference (MD) and 95% confidence interval (CI). Heterogeneity between studies was assessed by I2 tests. Sensitivity and subgroup analyses were also conducted. Results We included 16 RCTs comprising 3729 subjects in the meta-analyses. Anti-inflammatory therapies can significantly reduce the level of fasting plasma glucose (FPG) (MD = - 10.04; 95% CI: -17.69, - 2.40; P = 0.01), glycated haemoglobin (HbA1c) (MD = - 0.37; 95% CI: - 0.51, - 0.23; P < 0.00001), and C-reactive protein (CRP) (MD = - 1.05; 95% CI: - 1.50, - 0.60; P < 0.00001) compared with control, and therapies targeting IL-1β in combination with TNF-α have better effects on T2DM than targeting IL-1β or TNF-α alone. Subgroup analyses suggested that patients with short duration of T2DM may benefit more from anti-inflammatory therapies. Conclusion Our meta-analyses indicate that anti-inflammatory therapies targeting the pathogenic processes of diabetes can significantly reduce the level of FPG, HbA1c, and CRP in patients with T2DM.
Collapse
Affiliation(s)
- Dandan Li
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jiaxin Zhong
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qirui Zhang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jingjing Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- *Correspondence: Jingjing Zhang,
| |
Collapse
|
10
|
Ratnaningtyas NI, Hernayanti H, Ekowati N, Husen F. Ethanol extract of the mushroom Coprinus comatus exhibits antidiabetic and antioxidant activities in streptozotocin-induced diabetic rats. PHARMACEUTICAL BIOLOGY 2022; 60:1126-1136. [PMID: 35675226 PMCID: PMC9186368 DOI: 10.1080/13880209.2022.2074054] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/08/2022] [Accepted: 04/30/2022] [Indexed: 06/15/2023]
Abstract
CONTEXT Edible mushrooms have a long history of use in traditional Chinese or Japanese medicine. Coprinus comatus (O.F. Müll.) Pers. (Agaricaceae) contains antioxidant and antidiabetic agents. OBJECTIVE To identify the benefits of ethanol extracts of the C. comatus fruit body in streptozotocin-induced hyperglycaemic rats by evaluating their blood glucose, glycosylated haemoglobin (HbA1c), insulin, glucagon-like peptide-1 (GLP-1), dipeptidyl peptidase-4 (DPP-4), and glutathione (GSH) levels, with and without extract administration. MATERIALS AND METHODS Wistar rats were either left untreated or were administered 45 mg/kg body weight (BW) streptozotocin; 45 mg/kg BW metformin; or 250, 500, or 750 mg/kg BW extract for 14 days. The blood glucose, GLP-1, DPP-4, GSH, insulin, and HbA1c levels were determined. Data were analysed using analysis of variance and Duncan's multiple range tests. RESULTS Preliminary data showed that administration of C. comatus ethanol extract dose of 250, 500, and 750 mg orally has no toxicity effects after 24 h administration. The ethanolic extract of fruiting body of C. comatus considerably reduced the rat's fasting blood glucose levels 26.69%, and DPP-4 6.97% at dose of 750 mg. The extract reduced HbA1c 4-4.30%, increased GLP-1 71.09%, GSH 11.19% at dose of 500 mg, and increased insulin levels 13.83%. Extracts contain bioactive compounds such as flavonoid, alkaloid, terpenoids, vitamins C and E, rutin, and saponin. CONCLUSIONS The C. comatus extract can be used as herbal medicine that reduces diabetic symptoms. Further investigation on C. comatus extracts should be conducted with gas chromatography-mass spectrometry to characterise the bioactive compounds.
Collapse
Affiliation(s)
- Nuniek Ina Ratnaningtyas
- Department of Biology, Faculty of Biology, Jenderal Soedirman University, Central Java, Indonesia
| | - Hernayanti Hernayanti
- Department of Biology, Faculty of Biology, Jenderal Soedirman University, Central Java, Indonesia
| | - Nuraeni Ekowati
- Department of Biology, Faculty of Biology, Jenderal Soedirman University, Central Java, Indonesia
| | - Fajar Husen
- Department of Medical Laboratory of Technology, Bina Cipta Husada College of Health Science, Central Java, Indonesia
| |
Collapse
|
11
|
Andreone L, Fuertes F, Sétula C, Barcala Tabarrozzi AE, Orellano MS, Dewey RA, Bottino R, De Bosscher K, Perone MJ. Compound A attenuates proinflammatory cytokine-induced endoplasmic reticulum stress in beta cells and displays beneficial therapeutic effects in a mouse model of autoimmune diabetes. Cell Mol Life Sci 2022; 79:587. [PMID: 36370223 DOI: 10.1007/s00018-022-04615-5] [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: 07/22/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 11/15/2022]
Abstract
Type 1 diabetes (T1D) is characterized by an immune-mediated progressive destruction of the insulin-producing β-cells. Proinflammatory cytokines trigger endoplasmic reticulum (ER) stress and subsequent insulin secretory deficiency in cultured β-cells, mimicking the islet microenvironment in T1D. β-cells undergo physiologic ER stress due to the high rate of insulin production and secretion under stimulated conditions. Severe and uncompensated ER stress in β-cells is induced by several pathological mechanisms before onset and during T1D. We previously described that the small drug Compound A (CpdA), a selective glucocorticoid receptor (GR/NR3C1, nuclear receptor subfamily 3, group C, member 1) ligand with demonstrated inflammation-suppressive activity in vivo, is an effective modulator of effector T and dendritic cells and of macrophages, yet, in a GR-independent manner. Here, we focus on CpdA's therapeutic potential in T1D cellular and animal models. We demonstrate that CpdA improves the unfolded protein response (UPR) by attenuating ER stress and favoring the survival and function of β-cells exposed to an environment of proinflammatory cytokines. CpdA administration to NODscid mice adoptively transferred with diabetogenic splenocytes (from diabetic NOD mice) led to a delay of disease onset and reduction of diabetes incidence. Histological analysis of the pancreas showed a reduction in islet leukocyte infiltration (insulitis) and preservation of insulin expression in CpdA-treated normoglycemic mice in comparison with control group. These new findings together with our previous reports justify further studies on the administration of this small molecule as a novel therapeutic strategy with dual targets (effector immune and β-cells) during autoimmune diabetes.
Collapse
Affiliation(s)
- Luz Andreone
- Laboratory of Immuno-Endocrinology, Diabetes and Metabolism, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Pte. Perón 1500 (B1629AHJ), Pilar, Buenos Aires, Argentina
| | - Florencia Fuertes
- Laboratory of Immuno-Endocrinology, Diabetes and Metabolism, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Pte. Perón 1500 (B1629AHJ), Pilar, Buenos Aires, Argentina
| | - Carolina Sétula
- Laboratory of Immuno-Endocrinology, Diabetes and Metabolism, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Pte. Perón 1500 (B1629AHJ), Pilar, Buenos Aires, Argentina
| | - Andres E Barcala Tabarrozzi
- Laboratory of Immuno-Endocrinology, Diabetes and Metabolism, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Pte. Perón 1500 (B1629AHJ), Pilar, Buenos Aires, Argentina
| | - Miranda S Orellano
- Laboratory of Immuno-Endocrinology, Diabetes and Metabolism, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Pte. Perón 1500 (B1629AHJ), Pilar, Buenos Aires, Argentina
| | - Ricardo A Dewey
- Laboratorio de Terapia Génica Y Células Madre, Instituto Tecnológico de Chascomús (INTECH), CONICET-UNSAM, Buenos Aires, Argentina
| | - Rita Bottino
- Imagine Pharma, Pittsburgh, Pennsylvania, PA and Allegheny Health Network, Pittsburgh, PA, USA
| | - Karolien De Bosscher
- Receptor Research Laboratories, Nuclear Receptor Lab, VIB-Department of Medical Protein Research, VIB, Ghent University, Ghent, Belgium
| | - Marcelo J Perone
- Laboratory of Immuno-Endocrinology, Diabetes and Metabolism, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Pte. Perón 1500 (B1629AHJ), Pilar, Buenos Aires, Argentina.
| |
Collapse
|
12
|
Macrophages and neutrophils are necessary for ER stress-induced β cell loss. Cell Rep 2022; 40:111255. [PMID: 36001973 PMCID: PMC9444341 DOI: 10.1016/j.celrep.2022.111255] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 06/09/2022] [Accepted: 08/02/2022] [Indexed: 11/23/2022] Open
Abstract
Persistent endoplasmic reticulum (ER) stress induces islet inflammation and β cell loss. How islet inflammation contributes to β cell loss remains uncertain. We have reported previously that chronic overnutrition-induced ER stress in β cells causes Ripk3-mediated islet inflammation, macrophage recruitment, and a reduction of β cell numbers in a zebrafish model. We show here that β cell loss results from the intricate communications among β cells, macrophages, and neutrophils. Macrophage-derived Tnfa induces cxcl8a in β cells. Cxcl8a, in turn, attracts neutrophils to macrophage-contacted “hotspots” where β cell loss occurs. We also show potentiation of chemokine expression in stressed mammalian β cells by macrophage-derived TNFA. In Akita and db/db mice, there is an increase in CXCL15-positive β cells and intra-islet neutrophils. Blocking neutrophil recruitment in Akita mice preserves β cell mass and slows diabetes progression. These results reveal an important role of neutrophils in persistent ER stress-induced β cell loss. Yang et al. show a pivotal role of communications among β cells, macrophages, and neutrophils in chronic overnutrition-induced loss of pancreatic β cells in a diabetes-prone zebrafish model.
Collapse
|
13
|
Zhang X, Luo S, Wang M, Huang Q, Fang W, Li J, Liu T, Zhang Y, Deng Z, Liu CL, Guan S, Ayala JE, Flavell RA, Kulkarni RN, Libby P, Guo J, Liu Z, Shi GP. IL18 signaling causes islet β cell development and insulin secretion via different receptors on acinar and β cells. Dev Cell 2022; 57:1496-1511.e6. [DOI: 10.1016/j.devcel.2022.05.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 12/31/2021] [Accepted: 05/16/2022] [Indexed: 12/13/2022]
|
14
|
Li Y, Li T, Zhou Z, Xiao Y. Emerging roles of Galectin-3 in diabetes and diabetes complications: A snapshot. Rev Endocr Metab Disord 2022; 23:569-577. [PMID: 35083706 PMCID: PMC9156459 DOI: 10.1007/s11154-021-09704-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/13/2021] [Indexed: 12/13/2022]
Abstract
Galectin-3 is a member of the galectin family, widely expressed in immune cells and plays a role mainly in inflammation, autoimmunity, apoptosis, and chemotaxis. We summarized the roles of Galectin-3 in diabetes and its complications, as well as the underlying mechanisms. Clinical research has determined that the circulating level of Galectin-3 is closely related to diabetes and its complications, thus it is promising to use Galectin-3 as a predictor and biomarker for those diseases. Galectin-3 also may be considered as an ideal therapeutic target, which has broad prospects in the prevention and treatment of diabetes and its complications, especially macrovascular and microvascular complications.
Collapse
Affiliation(s)
- Yanhua Li
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, No. 139, Renmin Rd, Changsha, 410011, China
- Department of Metabolism and Endocrinology, The Third Hospital of Changsha, 176, West Labour Road, Changsha, 410011, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, No. 169 Changle West Rd, Xi'an, 710032, China.
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, No. 139, Renmin Rd, Changsha, 410011, China
| | - Yang Xiao
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, No. 139, Renmin Rd, Changsha, 410011, China.
| |
Collapse
|
15
|
Yuan S, Ye Z, Li Y, Zou J, Wu M, Wang K, Liao W, Shen J. Hypoglycemic Effect of Nobiletin via Regulation of Islet β-Cell Mitophagy and Gut Microbiota Homeostasis in Streptozocin-Challenged Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:5805-5818. [PMID: 35522926 DOI: 10.1021/acs.jafc.2c00148] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Nobiletin is a natural nutrient (or polymethoxyflavonoid) in orange peels exerting a preventive effect against metabolic diseases. However, there are very few reports on the hypoglycemic effect of nobiletin. In the present study, the hypoglycemic effect of nobiletin was investigated using NIT-1 cells and streptozocin (STZ)-challenged mouse models. Our results indicated that nobiletin could significantly suppress the high blood glucose in STZ-challenged mice. In addition, nobiletin could effectively activate the mitophagy and inhibit the inflammatory pathways in NIT-1 cells. The mitochondria membrane potential dysbiosis induced by glucotoxicity in NIT-1 cells was restored after treatment by nobiletin. Further investigation revealed that the hypoglycemic effect of nobiletin was mainly through regulation of gut microbiota dysbiosis, activation of mitophagy flux, inhibition of inflammasome expression, and restoration of islet morphological destruction in the pancreas of STZ-challenged mice. Our study revealed that nobiletin could be used as a functional food or drug candidate for the treatment of diabetes.
Collapse
Affiliation(s)
- Sijie Yuan
- Department of Endocrinology and Metabolic Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510630, China
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zichong Ye
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Ye Li
- Department of Endocrinology and Metabolic Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510630, China
| | - Jiaxuan Zou
- School of Biological Science, University of California Irvine, Irvine, California 92697, United States
| | - Mengting Wu
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Ke Wang
- Department of Food Science & Technology, National University of Singapore, Singapore 117542, Singapore
| | - Wenzhen Liao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jie Shen
- Department of Endocrinology and Metabolic Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510630, China
| |
Collapse
|
16
|
Akki R, Siracusa R, Cordaro M, Remigante A, Morabito R, Errami M, Marino A. Adaptation to oxidative stress at cellular and tissue level. Arch Physiol Biochem 2022; 128:521-531. [PMID: 31835914 DOI: 10.1080/13813455.2019.1702059] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Several in vitro and in vivo investigations have already proved that cells and tissues, when pre-exposed to low oxidative stress by different stimuli such as chemical, physical agents and environmental factors, display more resistance against subsequent stronger ischaemic injuries, resulting in an adaptive response known as ischaemic preconditioning (IPC). The aim of this review is to report the most recent knowledge about the complex adaptive mechanisms, including signalling transduction pathways, antioxidant systems, apoptotic and inflammation pathways, underlying cell protection against oxidative damage. In addition, an update about in vivo adaptation strategies in response to ischaemic/reperfusion episodes and brain trauma is also given.
Collapse
Affiliation(s)
- Rachid Akki
- Department of Biology, Faculty of Science, University of Abdelmalek Essaadi, Tetouan, Morocco
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Marika Cordaro
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Alessia Remigante
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Rossana Morabito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Mohammed Errami
- Department of Biology, Faculty of Science, University of Abdelmalek Essaadi, Tetouan, Morocco
| | - Angela Marino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| |
Collapse
|
17
|
Wang Z, Huang K, Xu J, Liu J, Zheng Y. Insights from Dysregulated mRNA Expression Profile of β-Cells in Response to Proinflammatory Cytokines. J Immunol Res 2022; 2022:4542487. [PMID: 35103245 PMCID: PMC8800623 DOI: 10.1155/2022/4542487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 12/28/2021] [Accepted: 01/08/2022] [Indexed: 12/17/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is a chronic autoimmune disease that is characterized by autoimmunity and its mediated β-cell damage. Chronic exposure of β-cells to proinflammatory cytokines is known to regulate the expression of many genes, subsequently resulting in the impairment of some signaling pathways involved with insulin production and secretion and/or β-cell apoptosis. In our study, RNA sequencing technology was applied to identify differentially expressed mRNAs in MIN6 cells treated with a mix of cytokines, including IL-1β, TNF-α, and IFN-γ. The results showed 809 upregulated and 946 downregulated protein-coding mRNAs in MIN6 cells upon the stimulation of cytokines. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) biological pathway analyses were performed to predict the functions of dysregulated genes. The networks of circRNA-mRNA were constructed between differentially mRNAs and dysregulated expressed circRNAs in our previous study. In addition, we selected 8 dysregulated mRNAs for further validation by quantitative real-time PCR. The RNA sequencing data showed 809 upregulated and 946 downregulated protein-coding mRNAs. GO analysis showed that the top 10 significant "biological processes," "cellular components," and "molecular functions" for upregulated mRNAs include "immune system process," "inflammatory response," and "innate immune response" and the top 10 for downregulated mRNAs include "cell cycle," "mitotic cytokinesis," and "cytoplasm." KEGG analysis showed that these differentially expressed genes were involved with "antigen processing and presentation," "TNF signaling pathway" and "type 1 diabetes," "cell cycle," "necroptosis," and "Rap1 signaling pathway." We also constructed the networks of differentially expressed circRNAs and mRNAs. We observed that upregulated circRNA 006029 and downregulated circRNA 000286 and 017277 were associated with the vast majority of selected dysregulated mRNAs, while circRNA 013053 was only related to the protein-coding gene, Slc7a2. To the summary, these data indicated that differentially expressed mRNAs may play key or partial roles in cytokine-mediated β-cell dysfunction and gave us the hint that circRNAs might regulate mRNAs, thereby contributing to the development of T1DM. The current study provided a systematic perspective on the potential functions and possible regulatory mechanisms of mRNAs in proinflammatory cytokine-induced β-cell destruction.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Changsha, Hunan 410011, China
- National Clinical Research Center for Metabolic Diseases, Changsha, Hunan 410011, China
| | - Kunlin Huang
- Center for Medical Research, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Jing Xu
- Department of Metabolism and Endocrinology, The First People's Hospital of Pingjiang, Pingjiang, Hunan 414500, China
| | - Jia Liu
- Center for Medical Research, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Ying Zheng
- Center for Medical Research, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
18
|
Brodnicki TC. A Role for lncRNAs in Regulating Inflammatory and Autoimmune Responses Underlying Type 1 Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1363:97-118. [DOI: 10.1007/978-3-030-92034-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
19
|
Azeredo CM, Cortese M, Munger KL, Ascherio A. Maternal prepregnancy BMI and physical activity and type 1 diabetes in the offspring. Pediatr Diabetes 2021; 22:992-1002. [PMID: 34260806 DOI: 10.1111/pedi.13248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 04/09/2021] [Accepted: 07/05/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Previous studies showed conflicting results on the association between maternal prepregnancy body mass index (BMI) and type 1 diabetes in the offspring, and the role of maternal prepregnancy physical activity is unclear. We aimed to assess whether maternal prepregnancy BMI and physical activity predict type 1 diabetes in their offspring. METHODS Prospective study including women participating in the Nurses' Health Study II with follow-up from 1989 to 2011. Women repeatedly reported their BMI and physical activity, from which prepregnancy exposures were derived; and retrospectively reported their BMI at age 18 and physical activity at ages 18-22, considered early adulthood exposure. We estimated risk ratios (RR) and 95% confidence intervals (95%CI) using generalized estimating equations, adjusted for covariates. Findings at p < 0.05 were considered statistically significant. RESULTS We identified 276 cases of type 1 diabetes among offspring (n = 70,168) with maternal prepregnancy information and 448 cases among offspring (n = 111,692) with maternal early adulthood information. Prepregnancy and early adulthood maternal BMI and physical activity were not associated with offspring type 1 diabetes. The RR comparing overweight to normal weight mothers was 1.08 (95%CI: 0.73-1.59) and comparing obese to normal weight was 0.94 (95%CI: 0.49-1.79, p-trend: 0.98). Comparing highest to lowest quartile of maternal physical activity the RR was 0.90 (95%CI: 0.61-1.32; p-trend: 0.73). Maternal type 2 diabetes was associated with an increased risk of type 1 diabetes in the offspring (RR = 1.87; 95%CI: 1.25-2.80). CONCLUSIONS Our findings do not support a relationship between maternal prepregnancy BMI or physical activity and the risk of type 1 diabetes in the offspring.
Collapse
Affiliation(s)
- Catarina Machado Azeredo
- School of Medicine, Federal University of Uberlandia, Uberlandia, Minas Gerais, Brazil.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Marianna Cortese
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Kassandra L Munger
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Alberto Ascherio
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Ding F, Luo X, Tu Y, Duan X, Liu J, Jia L, Zheng P. Alpk1 Sensitizes Pancreatic Beta Cells to Cytokine-Induced Apoptosis via Upregulating TNF-α Signaling Pathway. Front Immunol 2021; 12:705751. [PMID: 34621265 PMCID: PMC8490819 DOI: 10.3389/fimmu.2021.705751] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 09/06/2021] [Indexed: 11/13/2022] Open
Abstract
Pancreatic beta cell failure is the hallmark of type 1 diabetes (T1D). Recent studies have suggested that pathogen recognizing receptors (PRRs) are involved in the survival, proliferation and function of pancreatic beta cells. So far, little is known about the role of alpha-protein kinase 1 (ALPK1), a newly identified cytosolic PRR specific for ADP-β-D-manno-heptose (ADP-heptose), in beta cell survival. In current study we aimed to fill the knowledge gap by investigating the role of Alpk1 in the apoptosis of MIN6 cells, a murine pancreatic beta cell line. We found that the expression of Alpk1 was significantly elevated in MIN6 cells exposed to pro-inflammatory cytokines, but not to streptozotocin, low-dose or high-dose glucose. Activation of Alpk1 by ADP heptose alone was insufficient to induce beta cell apoptosis. However, it significantly exacerbated cytokine-induced apoptosis in MIN6 cells. Mechanistic investigations showed that Alpk1 activation was potent to further induce the expression of tumor necrosis factor (TNF)-α and Fas after cytokine stimulation, possibly due to enhanced activation of the TIFA/TAK1/NF-κB signaling axis. Treatment of GLP-1 receptor agonist decreased the expression of TNF-α and Fas and improved the survival of beta cells exposed to pro-inflammatory cytokines and ADP heptose. In summary, our data suggest that Alpk1 sensitizes beta cells to cytokine-induced apoptosis by potentiating TNF-α signaling pathway, which may provide novel insight into beta cell failure and T1D development.
Collapse
Affiliation(s)
- Fei Ding
- Department of Endocrinology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Xi Luo
- Department of Endocrinology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Yiting Tu
- Department of Neurology, Shenzhen Samii International Medical Center (The Fourth People's Hospital of Shenzhen), Shenzhen, China
| | - Xianlan Duan
- Department of Endocrinology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Jia Liu
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Lijing Jia
- Department of Endocrinology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Peilin Zheng
- Department of Endocrinology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
21
|
Martorell M, Castro N, Victoriano M, Capó X, Tejada S, Vitalini S, Pezzani R, Sureda A. An Update of Anthraquinone Derivatives Emodin, Diacerein, and Catenarin in Diabetes. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:3313419. [PMID: 34589130 PMCID: PMC8476274 DOI: 10.1155/2021/3313419] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/09/2021] [Indexed: 12/15/2022]
Abstract
Diabetes is part of metabolic diseases and is characterized by high blood sugar levels over a prolonged period as result of an insulin-deficient production or an inappropriate response to insulin by our cells. This chronic disease was the direct cause of 1.6 million deaths in 2016 as reported by the World Health Organization. Emodin is a natural product and active ingredient of various Chinese herbs with the chemical formula 1,3,8-trihydroxy-6-methylanthraquinone. Diacerein is another naturally occurring anthraquinone (1,8-diacetoxy-3-carboxyanthraquinone) commonly used as commercial drug to treat osteoarthritis. These two anthraquinone derivatives have been shown to exert antidiabetic activities. Emodin seems to enhance the glucose tolerance and insulin sensibility via activation of PPARγ and modulation of metabolic-related genes. Diacerein seems to decrease inflammatory cytokines and increase insulin secretion enhancing insulin sensibility and therefore improving glucose control. Other naturally occurring anthraquinone derivatives, such as catenarin (1,4,6,8-tetrahydroxy-3-methylanthraquinone), have been shown to have antidiabetic activities although few studies have been performed. The synthesis of new emodin derivatives is increasing, but these new molecules have not been tested for diabetes treatment. In the current work, available literature on anthraquinone derivatives' effects in diabetes disease is reviewed. Moreover, we discuss the chemistry, food sources, bioavailability, and toxicity of the naturally occurring anthraquinone with antidiabetic effects.
Collapse
Affiliation(s)
- Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, University of Concepción, Concepción 4070386, Chile
- Centre for Healthy Living, University of Concepción, Concepción 4070386, Chile
| | - Natalia Castro
- Department of Nutrition and Dietetics, Faculty of Pharmacy, University of Concepción, Concepción 4070386, Chile
| | - Montserrat Victoriano
- Department of Nutrition and Dietetics, Faculty of Pharmacy, University of Concepción, Concepción 4070386, Chile
| | - Xavier Capó
- Research Group in Community Nutrition and Oxidative Stress, University Research Institute of Health Sciences (IUNICS), University of the Balearic Islands, Palma de Mallorca 07122, Spain
| | - Silvia Tejada
- Laboratory of Neurophysiology, Department of Biology, University Research Institute of Health Sciences (IUNICS), University of Balearic Islands, Palma E-07122, Balearic Islands, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, Madrid E-28029, Spain
- Research Institute of the Balearic Islands, Palma de Mallorca E-07120, Spain
| | - Sara Vitalini
- Department of Agricultural and Environmental Sciences, Università degli Studi di Milano, Via G. Celoria 2 20133, Milan, Italy
| | - Raffaele Pezzani
- Phytotherapy Lab (PhT-Lab), Endocrinology Unit, Department of Medicine (DIMED), University of Padova, via Ospedale 105, Padova 35128, Italy
- AIROB, Associazione Italiana per la Ricerca Oncologica di Base, Padova, Italy
| | - Antoni Sureda
- Research Group in Community Nutrition and Oxidative Stress, University Research Institute of Health Sciences (IUNICS), University of the Balearic Islands, Palma de Mallorca 07122, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, Madrid E-28029, Spain
- Research Institute of the Balearic Islands, Palma de Mallorca E-07120, Spain
| |
Collapse
|
22
|
Divalent Metal Transporter 1 Knock-Down Modulates IL-1β Mediated Pancreatic Beta-Cell Pro-Apoptotic Signaling Pathways through the Autophagic Machinery. Int J Mol Sci 2021; 22:ijms22158013. [PMID: 34360779 PMCID: PMC8348373 DOI: 10.3390/ijms22158013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/23/2021] [Accepted: 07/24/2021] [Indexed: 12/21/2022] Open
Abstract
Pro-inflammatory cytokines promote cellular iron-import through enhanced divalent metal transporter-1 (DMT1) expression in pancreatic β-cells, consequently cell death. Inhibition of β-cell iron-import by DMT1 silencing protects against apoptosis in animal models of diabetes. However, how alterations of signaling networks contribute to the protective action of DMT1 knock-down is unknown. Here, we performed phosphoproteomics using our sequential enrichment strategy of mRNA, protein, and phosphopeptides, which enabled us to explore the concurrent molecular events in the same set of wildtype and DMT1-silenced β-cells during IL-1β exposure. Our findings reveal new phosphosites in the IL-1β-induced proteins that are clearly reverted by DMT1 silencing towards their steady-state levels. We validated the levels of five novel phosphosites of the potential protective proteins using parallel reaction monitoring. We also confirmed the inactivation of autophagic flux that may be relevant for cell survival induced by DMT1 silencing during IL-1β exposure. Additionally, the potential protective proteins induced by DMT1 silencing were related to insulin secretion that may lead to improving β-cell functions upon exposure to IL-1β. This global profiling has shed light on the signal transduction pathways driving the protection against inflammation-induced cell death in β-cells after DMT1 silencing.
Collapse
|
23
|
Wang Z, Deng C, Zheng Y. Involvement of circRNAs in Proinflammatory Cytokines-Mediated β-Cell Dysfunction. Mediators Inflamm 2021; 2021:5566453. [PMID: 34054343 PMCID: PMC8112919 DOI: 10.1155/2021/5566453] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/11/2021] [Accepted: 04/22/2021] [Indexed: 01/25/2023] Open
Abstract
AIM During the initial stage of type 1 diabetes, prolonged exposure of pancreatic β-cell to proinflammatory cytokines such as IL-1β, TNF-α, and IFN-γ results in a decreased capacity to produce and release insulin, as well as cell loss by apoptosis. Circular RNAs (circRNAs) are a new class of endogenous noncoding RNAs (ncRNAs) with closed loop with no free ends. circRNAs have been reported to be participated in the development of many diseases. As little is known about their role in insulin-secreting cells, this study is aimed at evaluating their contribution in the process of inflammation-induced β-cell damage. METHODS circRNA expression profile of MIN6 cells stimulated with a mix of cytokines, including IL-1β, IFN-γ, and TNF-α, was detected by circRNA microarrays. Four dysregulated circRNAs were validated by qRT-PCR. The involvement of the selected circRNAs in β-cell dysfunction was tested after their inhibition in MIN6 cells. MicroRNA target prediction software and multiple bioinformatic approaches were used to predict the targeting genes of circRNAs and analyze possible functions of the circRNAs. RESULTS 1020 upregulated and 902 downregulated circRNAs were identified in cytokines-treated β-cells. Inhibition of circRNAs 000286 and 017277 in β-cells could promote β-cell apoptosis and affect insulin biosynthesis and secretion. GO analysis enriched terms such as regulation of transcription and regulation of gene expression and KEGG analysis enriched top pathways included TGF-β and MAPK signaling pathways. CONCLUSIONS The data shows that circRNAs may be involved in proinflammatory cytokines-mediated β-cell dysfunction and suggests the involvement of circRNAs in the development of type 1 diabetes.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, 410011 Hunan, China
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, 410011 Hunan, China
| | - Chao Deng
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, 410011 Hunan, China
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, 410011 Hunan, China
| | - Ying Zheng
- Center for Medical Research, The Second Xiangya Hospital, Central South University, Changsha, 410011 Hunan, China
| |
Collapse
|
24
|
Merjaneh L, Toprak D, McNamara S, Nay L, Sullivan E, Rosenfeld M. Acute hyperglycaemia in cystic fibrosis pulmonary exacerbations. Endocrinol Diabetes Metab 2021; 4:e00208. [PMID: 33855211 PMCID: PMC8029509 DOI: 10.1002/edm2.208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/27/2020] [Accepted: 11/08/2020] [Indexed: 11/07/2022] Open
Abstract
Background Hyperglycaemia may contribute to failure to recover from pulmonary exacerbations in cystic fibrosis (CF). We aimed to evaluate the prevalence and mechanism of hyperglycaemia during and post-exacerbations. Methods Nine paediatric CF patients, not on insulin, hospitalized for intravenous antibiotics, underwent an oral glucose tolerance test (OGTT) and continuous glucose monitoring (CGM) upon admission (visit 1) and an OGTT 2 weeks (visit 2) and 6 weeks to 12 months later when at stable baseline (visit 3). Insulin and glucose levels were measured before, 30, 60 and 120 min after glucose ingestion during OGTT. Hyperglycaemia on OGTT was defined according to the American Diabetes Association criteria as abnormal OGTT or consistent with diabetes. Hyperglycaemia on CGM was defined as CGM time above 140 mg/dL > 4.5%. Results At visit 1, 8/9 patients had hyperglycaemia on both CGM and OGTT (2 diabetes and 6 abnormal OGTT). At visit 2, 5/8 had hyperglycaemia (all abnormal OGTT). At visit 3, (median (IQR) time since visit 1, 4.9 (3.8-6.3) months), 5/7 had hyperglycaemia (2 diabetes and 3 abnormal OGTT). At visits 1, 2 and 3, respectively, mean (SD) 2-hour OGTT glucose was 175.8 (42.3), 146.3 (31.9) and 176.9 (51.7) mg/dL. CGM time above 140 mg/dL at visit 1 was 25.3% (16.9). Insulin AUC decreased from visit 2 (median (IQR) 5449 (3321-8123) mcIU-min/mL) to visit 3 (3234 (2913-3680) mcIU-min/mL). Conclusion Hyperglycaemia is prevalent during paediatric CF exacerbations; it appears to improve with exacerbation treatment but to worsen later in association with decreased insulin secretion.
Collapse
Affiliation(s)
- Lina Merjaneh
- Division of Endocrinology and DiabetesSeattle Children’s HospitalSeattleWAUSA
- Department of PediatricsUniversity of WashingtonSeattleWAUSA
| | - Demet Toprak
- Department of PediatricsUniversity of WashingtonSeattleWAUSA
- Division of Pulmonary and Sleep MedicineSeattle Children’s HospitalSeattleWAUSA
| | - Sharon McNamara
- Division of Pulmonary and Sleep MedicineSeattle Children’s HospitalSeattleWAUSA
| | - Laura Nay
- Division of Pulmonary and Sleep MedicineSeattle Children’s HospitalSeattleWAUSA
| | - Erin Sullivan
- Children’s Core for Biomedical StatisticsCenter for Clinical and Translational ResearchSeattle Children’s HospitalSeattleWAUSA
| | - Margaret Rosenfeld
- Department of PediatricsUniversity of WashingtonSeattleWAUSA
- Division of Pulmonary and Sleep MedicineSeattle Children’s HospitalSeattleWAUSA
| |
Collapse
|
25
|
Ge Y, Li B, Yang Y, Feng C, Tang X, Shi Y, Le G, Sun J. Oxidized Pork Induces Disorders of Glucose Metabolism in Mice. Mol Nutr Food Res 2021; 65:e2000859. [PMID: 33502107 DOI: 10.1002/mnfr.202000859] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/21/2020] [Indexed: 02/27/2024]
Abstract
SCOPE Consumption of red meat, particularly processed red meat, has been reported to be associated with type 2 diabetes risk, and oxidized proteins and amino acids may be involved in this process. This study explores the effects of pork with varying degrees of oxidative injury caused by cooking on glucose metabolism in mice. METHODS AND RESULTS Cooked pork is freeze-dried to prepare animal feed. Mice are fed either a control diet (CON), a low- (LOP), or a high-oxidative injury pork diet (HOP) for 12 weeks. Intake of HOP causes hyperglycemia, hypoinsulinemia, and impaired glucose tolerance, indicating a glucose metabolism disorder. Accumulation of oxidation products increases oxidative stress and inflammatory response, which impairs pancreatic islet β cells function and reduces insulin secretion. Moreover, HOP-mediated hyperglycemia can be partly attributed to elevated hepatic glucose output, as indicated by increased gluconeogenesis and glycogenolysis, and decreased glycolysis and glycogen content. Changes in these processes may be regulated by reduced insulin levels and suppression of the insulin receptor substrate-1 (IRS-1)/phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway and its downstream signaling molecules. CONCLUSION HOP intake induces disorders of glucose metabolism by impairing pancreatic insulin secretion and increasing hepatic glucose output. Protein oxidation plays a key role in abnormal glucose metabolism induced by HOP.
Collapse
Affiliation(s)
- Yueting Ge
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Bowen Li
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Yuhui Yang
- College of Grain and Food Science, Henan University of Technology, Zhengzhou, 450001, China
| | - Chuanxing Feng
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Xue Tang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Yonghui Shi
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Guowei Le
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Jin Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| |
Collapse
|
26
|
Al-Absi B, AL-Habori M, Saif-Ali R. Plasma Lipocalin-2 and Adiponectin are Affected by Obesity Rather Than Type 2 Diabetes Mellitus per se. Diabetes Metab Syndr Obes 2021; 14:4547-4556. [PMID: 34815681 PMCID: PMC8605802 DOI: 10.2147/dmso.s338254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/03/2021] [Indexed: 04/20/2023] Open
Abstract
PURPOSE Changes in plasma adipocytokines and inflammatory markers in type 2 DM remain controversial as to whether they are due to obesity or directly associated with the diabetic state. Our objective was to study the effect of obesity and diabetes on plasma lipocalin-2 (LCN2), adiponectin, and interleukin-1β (IL-1β) by comparing their levels in non-diabetic obese subjects and non-obese type 2 DM patients, as well as determining the association of these adipocytokines with metabolic syndrome factors and diabetic parameters. PATIENTS AND METHODS In this study, 85 Yemeni male volunteers aged 30-60 years old were enrolled, 25 of whom were healthy subjects with BMI < 25 kg/m2 served as control; 30 non-diabetic obese subjects (BMI ≥ 30 kg/m2 and FBG < 6.1 mmol/l); and 30 non-obese type 2 DM patients (BMI < 25 kg/m2 and FBG > 7 mmol/l). RESULTS Lipocalin-2 and adiponectin were significantly (p = 0.043 and p = 0.034) lower in non-diabetic obese subjects by 16.2% and 29.7% with respect to control group, with no effect in the non-obese type 2 DM patients. Moreover, LCN2 was significantly (p = 0.04) lower in the non-diabetic obese subjects by 15.8% as compared with the non-obese type 2 DM patients, with no significant difference in adiponectin levels. In contrast, serum IL-1β was significantly (p = 0.001 and p = 0.003) higher in both non-diabetic obese subjects and the non-obese type 2 DM patients by 76.5% and 67.7% as compared to control group. The significant decrease in both LCN2 and adiponectin and the significant increase in IL-1β in the non-diabetic obese subjects disappeared upon adjustment for waist circumference (WC). In contrast, the significant increase in IL-1β in the non-obese Type 2 DM patients was not affected upon adjustment for WC. CONCLUSION Plasma LCN2 and adiponectin were not affected by diabetes per se, suggesting that the observed changes in LCN2 and adiponectin in type 2 DM may be due to obesity rather than the diabetic state, whereas IL-1β levels were affected by both obesity and diabetes.
Collapse
Affiliation(s)
- Boshra Al-Absi
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Health Sciences, University of Sana`a, Sana`a, Yemen
| | - Molham AL-Habori
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Health Sciences, University of Sana`a, Sana`a, Yemen
- Correspondence: Molham AL-Habori Email
| | - Riyadh Saif-Ali
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Health Sciences, University of Sana`a, Sana`a, Yemen
| |
Collapse
|
27
|
Pyrillou K, Burzynski LC, Clarke MCH. Alternative Pathways of IL-1 Activation, and Its Role in Health and Disease. Front Immunol 2020; 11:613170. [PMID: 33391283 PMCID: PMC7775495 DOI: 10.3389/fimmu.2020.613170] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023] Open
Abstract
Cytokines activate or inhibit immune cell behavior and are thus integral to all immune responses. IL-1α and IL-1β are powerful apical cytokines that instigate multiple downstream processes to affect both innate and adaptive immunity. Multiple studies show that IL-1β is typically activated in macrophages after inflammasome sensing of infection or danger, leading to caspase-1 processing of IL-1β and its release. However, many alternative mechanisms activate IL-1α and IL-1β in atypical cell types, and IL-1 function is also important for homeostatic processes that maintain a physiological state. This review focuses on the less studied, yet arguably more interesting biology of IL-1. We detail the production by, and effects of IL-1 on specific innate and adaptive immune cells, report how IL-1 is required for barrier function at multiple sites, and discuss how perturbation of IL-1 pathways can drive disease. Thus, although IL-1 is primarily studied for driving inflammation after release from macrophages, it is clear that it has a multifaceted role that extends far beyond this, with various unconventional effects of IL-1 vital for health. However, much is still unknown, and a detailed understanding of cell-type and context-dependent actions of IL-1 is required to truly understand this enigmatic cytokine, and safely deploy therapeutics for the betterment of human health.
Collapse
Affiliation(s)
| | | | - Murray C. H. Clarke
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| |
Collapse
|
28
|
Abstract
Imeglimin is the first of the "glimins," a new class of drugs developed for the treatment of type 2 diabetes mellitus (T2DM). This review highlights its mechanism of action and its context in the field of T2DM treatment. Preclinical data in multiple rodent models have detailed significant effects on mitochondria, particularly improved mitochondrial bioenergetics. This includes changes favoring complex II and complex III metabolism, a mechanism potentially promoting increased fatty acid oxidation, leading to the decrease in hepatic lipid accumulation observed in these mice. Imeglimin was also shown to increase muscle glucose uptake and decrease hepatic glucose production, both in vitro and in vivo. Though studies have also shown imeglimin to significantly improve insulin secretion and decrease β-cell death, whether its physiologic effects are purely insulin dependent remains unclear. Early preclinical studies have shown evidence for improvements in cardiac and renal function in rats with metabolic syndrome, effects not conferred by most currently available T2DM drugs. Clinical studies of imeglimin in humans have shown increased insulin secretion, along with decreased fasting plasma glucose and glycated hemoglobin. Its observed efficacy was comparable to that of currently available agents metformin and sitagliptin and was increased when given in combination with either agent. When considered alongside its benign safety profile reported in patients with chronic kidney disease, imeglimin shows true promise to provide a novel mechanism for T2DM treatment, with potential application in a larger, more comprehensive patient population.
Collapse
|
29
|
Wallia A, Prince G, Touma E, El Muayed M, Seley JJ. Caring for Hospitalized Patients with Diabetes Mellitus, Hyperglycemia, and COVID-19: Bridging the Remaining Knowledge Gaps. Curr Diab Rep 2020; 20:77. [PMID: 33244614 PMCID: PMC7690847 DOI: 10.1007/s11892-020-01366-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/26/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW This review discusses the interplay between coronavirus disease 2019 (COVID-19, caused by SARS-CoV-2 infection), diabetes mellitus, and hyperglycemia in the hospital setting. There are data emerging about diabetes and hyperglycemia, their prevalence, and potential risks in the setting of SARS-CoV-2 infection and COVID-19. RECENT FINDINGS It is known that viral infections exert effects on beta cell function and insulin resistance. Therefore, much can be learned about SARS-CoV-2/COVID-19 from examining these known relationships. Such pathophysiological underpinnings may unlock greater understanding as we navigate atypical cases of hyperglycemia, severe insulin resistance, and diabetic ketoacidosis amidst COVID-19. Glycemic outcomes likely have beneficial effects on morbidity and mortality, but this needs to be studied. Changes in diabetes-related protocols and new technology can be deployed in the inpatient setting to potentially improve healthcare worker and patient safety; however, one must weigh the risks and benefits of implementation during a pandemic. Ultimately, knowledge and research must be shared at record speed to combat this global crisis.
Collapse
Affiliation(s)
- Amisha Wallia
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Institute of Public Health, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Grace Prince
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Emilie Touma
- Institute of Public Health, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Malek El Muayed
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jane Jeffrie Seley
- Division of Endocrinology, Diabetes, and Metabolism, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
30
|
Giri KR, de Beaurepaire L, Jegou D, Lavy M, Mosser M, Dupont A, Fleurisson R, Dubreil L, Collot M, Van Endert P, Bach JM, Mignot G, Bosch S. Molecular and Functional Diversity of Distinct Subpopulations of the Stressed Insulin-Secreting Cell's Vesiculome. Front Immunol 2020; 11:1814. [PMID: 33101266 PMCID: PMC7556286 DOI: 10.3389/fimmu.2020.01814] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/07/2020] [Indexed: 12/15/2022] Open
Abstract
Beta cell failure and apoptosis following islet inflammation have been associated with autoimmune type 1 diabetes pathogenesis. As conveyors of biological active material, extracellular vesicles (EV) act as mediators in communication with immune effectors fostering the idea that EV from inflamed beta cells may contribute to autoimmunity. Evidence accumulates that beta exosomes promote diabetogenic responses, but relative contributions of larger vesicles as well as variations in the composition of the beta cell's vesiculome due to environmental changes have not been explored yet. Here, we made side-by-side comparisons of the phenotype and function of apoptotic bodies (AB), microvesicles (MV) and small EV (sEV) isolated from an equal amount of MIN6 beta cells exposed to inflammatory, hypoxic or genotoxic stressors. Under normal conditions, large vesicles represent 93% of the volume, but only 2% of the number of the vesicles. Our data reveal a consistently higher release of AB and sEV and to a lesser extent of MV, exclusively under inflammatory conditions commensurate with a 4-fold increase in the total volume of the vesiculome and enhanced export of immune-stimulatory material including the autoantigen insulin, microRNA, and cytokines. Whilst inflammation does not change the concentration of insulin inside the EV, specific Toll-like receptor-binding microRNA sequences preferentially partition into sEV. Exposure to inflammatory stress engenders drastic increases in the expression of monocyte chemoattractant protein 1 in all EV and of interleukin-27 solely in AB suggesting selective sorting toward EV subspecies. Functional in vitro assays in mouse dendritic cells and macrophages reveal further differences in the aptitude of EV to modulate expression of cytokines and maturation markers. These findings highlight the different quantitative and qualitative imprints of environmental changes in subpopulations of beta EV that may contribute to the spread of inflammation and sustained immune cell recruitment at the inception of the (auto-) immune response.
Collapse
Affiliation(s)
| | | | | | - Margot Lavy
- IECM, ONIRIS, INRAE, USC1383, Nantes, France
| | | | - Aurelien Dupont
- MRic, Biosit, UMS3480 CNRS, University of Rennes 1, Rennes, France
| | | | - Laurence Dubreil
- PAnTher, INRAE, Oniris, Université Bretagne Loire, Nantes, France
| | - Mayeul Collot
- Laboratoire de Biophotonique et Pharmacologie, UMR CNRS 7213, Université de Strasbourg, Illkirch, France
| | - Peter Van Endert
- Université Paris Descartes, Paris, France.,INSERM, U1151, Institut Necker-Enfants Malades, Paris, France
| | | | | | | |
Collapse
|
31
|
Abstract
Diabetes mellitus (DM) is a complication of chronic pancreatitis (CP). Whether pancreatogenic diabetes associated with CP-DM represents a discrete pathophysiologic entity from type 2 DM (T2DM) remains uncertain. Addressing this question is needed for development of specific measures to manage CP-DM. We approached this question from a unique standpoint, hypothesizing that if CP-DM and T2DM are separate disorders, they should be genetically distinct. To test this hypothesis, we sought to determine whether a genetic risk score (GRS) based on validated single nucleotide polymorphisms for T2DM could distinguish between groups with CP-DM and T2DM.
Collapse
|
32
|
Frezza C. Publisher Correction: A BAD portion of glucose can be good for inflamed beta cells. Nat Metab 2020; 2:649. [PMID: 32694790 PMCID: PMC7115866 DOI: 10.1038/s42255-020-0236-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
Collapse
|
33
|
Hypomethylation of IL1RN and NFKB1 genes is linked to the dysbalance in IL1β/IL-1Ra axis in female patients with type 2 diabetes mellitus. PLoS One 2020; 15:e0233737. [PMID: 32470060 PMCID: PMC7259508 DOI: 10.1371/journal.pone.0233737] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023] Open
Abstract
Inflammation has received considerable attention in the pathogenesis of type 2 diabetes mellitus (T2DM). Supporting this concept, enhanced expression of interleukin (IL)-1β and increased infiltration of macrophages are observed in pancreatic islets of patients with T2DM. Although IL-1 receptor antagonist (IL-1Ra) plays a major role in controlling of IL-1β-mediated inflammation, its counteraction effects and epigenetic alterations in T2DM are less studied. Thus, we aimed to analyze the DNA methylation status in IL1RN, RELA (p65) and NFKB1 (p50) genes in peripheral blood mononuclear cells (PBMCs) from treated T2DM patients (n = 35) and age-/sex- matched healthy controls (n = 31). Production of IL-1β and IL-1Ra was analyzed in plasma and supernatants from LPS-induced PBMCs. Immunomodulatory effects of IL-1β and IL-1Ra were studied on THP-1 cells. Average DNA methylation level of IL1RN and NFKB1 gene promoters was significantly decreased in T2DM patients in comparison with healthy controls (P< 0.05), which was associated with the increased IL-1Ra (P< 0.001) and IL-1β (P = 0.039) plasma levels in T2DM patients. Negative association between average methylation of IL1RN gene and IL-1Ra plasma levels were observed in female T2DM patients. Methylation of NFKB1 gene was negatively correlated with IL-1Ra levels in the patients and positively with IL-1β levels in female patients. LPS-stimulated PBMCs from female patients failed to raise IL-1β production, while the cells from healthy females increased IL-1β production in comparison with unstimulated cells (P< 0.001). Taken together, the findings suggest that hypomethylation of IL1RN and NFKB1 gene promoters may promote the increased IL-1β/IL-1Ra production and regulate chronic inflammation in T2DM. Further studies are necessary to elucidate the causal direction of these associations and potential role of IL-1Ra in anti-inflammatory processes in treated patients with T2DM.
Collapse
|
34
|
Frezza C. A BAD portion of glucose can be good for inflamed beta cells. Nat Metab 2020; 2:383-384. [PMID: 32694661 DOI: 10.1038/s42255-020-0198-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
35
|
Ying W, Fu W, Lee YS, Olefsky JM. The role of macrophages in obesity-associated islet inflammation and β-cell abnormalities. Nat Rev Endocrinol 2020; 16:81-90. [PMID: 31836875 PMCID: PMC8315273 DOI: 10.1038/s41574-019-0286-3] [Citation(s) in RCA: 206] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/24/2019] [Indexed: 12/16/2022]
Abstract
Chronic, unresolved tissue inflammation is a well-described feature of obesity, type 2 diabetes mellitus (T2DM) and other insulin-resistant states. In this context, adipose tissue and liver inflammation have been particularly well studied; however, abundant evidence demonstrates that inflammatory processes are also activated in pancreatic islets from obese animals and humans with obesity and/or T2DM. In this Review, we focus on the characteristics of immune cell-mediated inflammation in islets and the consequences of this with respect to β-cell function. In contrast to type 1 diabetes mellitus, the dominant immune cell type causing inflammation in obese and T2DM islets is the macrophage. The increased macrophage accumulation in T2DM islets primarily arises through local proliferation of resident macrophages, which then provide signals (such as platelet-derived growth factor) that drive β-cell hyperplasia (a classic feature of obesity). In addition, islet macrophages also impair the insulin secretory capacity of β-cells. Through these mechanisms, islet-resident macrophages underlie the inflammatory response in obesity and mechanistically participate in the β-cell hyperplasia and dysfunction that characterizes this insulin-resistant state. These findings point to the possibility of therapeutics that target islet inflammation to elicit beneficial effects on β-cell function and glycaemia.
Collapse
Affiliation(s)
- Wei Ying
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Wenxian Fu
- Pediatric Diabetes Research Center, Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Yun Sok Lee
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jerrold M Olefsky
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
36
|
Zatterale F, Longo M, Naderi J, Raciti GA, Desiderio A, Miele C, Beguinot F. Chronic Adipose Tissue Inflammation Linking Obesity to Insulin Resistance and Type 2 Diabetes. Front Physiol 2020; 10:1607. [PMID: 32063863 PMCID: PMC7000657 DOI: 10.3389/fphys.2019.01607] [Citation(s) in RCA: 515] [Impact Index Per Article: 128.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 12/23/2019] [Indexed: 12/13/2022] Open
Abstract
Obesity is one of the major health burdens of the 21st century as it contributes to the growing prevalence of its related comorbidities, including insulin resistance and type 2 diabetes. Growing evidence suggests a critical role for overnutrition in the development of low-grade inflammation. Specifically, chronic inflammation in adipose tissue is considered a crucial risk factor for the development of insulin resistance and type 2 diabetes in obese individuals. The triggers for adipose tissue inflammation are still poorly defined. However, obesity-induced adipose tissue expansion provides a plethora of intrinsic signals (e.g., adipocyte death, hypoxia, and mechanical stress) capable of initiating the inflammatory response. Immune dysregulation in adipose tissue of obese subjects results in a chronic low-grade inflammation characterized by increased infiltration and activation of innate and adaptive immune cells. Macrophages are the most abundant innate immune cells infiltrating and accumulating into adipose tissue of obese individuals; they constitute up to 40% of all adipose tissue cells in obesity. In obesity, adipose tissue macrophages are polarized into pro-inflammatory M1 macrophages and secrete many pro-inflammatory cytokines capable of impairing insulin signaling, therefore promoting the progression of insulin resistance. Besides macrophages, many other immune cells (e.g., dendritic cells, mast cells, neutrophils, B cells, and T cells) reside in adipose tissue during obesity, playing a key role in the development of adipose tissue inflammation and insulin resistance. The association of obesity, adipose tissue inflammation, and metabolic diseases makes inflammatory pathways an appealing target for the treatment of obesity-related metabolic complications. In this review, we summarize the molecular mechanisms responsible for the obesity-induced adipose tissue inflammation and progression toward obesity-associated comorbidities and highlight the current therapeutic strategies.
Collapse
Affiliation(s)
- Federica Zatterale
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Michele Longo
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Jamal Naderi
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Environmental, Biological, and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Gregory Alexander Raciti
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Antonella Desiderio
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Claudia Miele
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Francesco Beguinot
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| |
Collapse
|
37
|
Ma L, Su H, Wang Y, Zhou Y, Kang Z, Xu Y, Gao J. Interleukin-1β (IL-1β) C-511T polymorphism is associated with susceptibility to coronary artery disease in type 2 diabetic patients. EUR J INFLAMM 2020. [DOI: 10.1177/2058739220918047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Variants of the interleukin-1β (IL-1β) gene have been associated with type 2 diabetes (T2D) and coronary artery diseases (CAD). However, association of IL-1β polymorphisms with diabetic patients having CAD clinical manifestation has not been studied yet. In this study, we aim to decipher the role of IL-1β common promoter variants with susceptibility/resistance to development of CAD in T2D patients. T2D patients with (n = 134) or without CAD (n = 533) were enrolled. A total of 513 essentially healthy individuals from the same population were included in this study as control. Plasma levels of IL-1β were quantified by enzyme-linked immunosorbent assay (ELISA) kit as per instructions from the manufacturer. IL-1β promoter variants (T-31C and C-511T) were genotyped by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). T2D patients displayed higher level of plasma IL-1β in comparison to healthy controls. Prevalence of variants for IL-1β (C-511T) polymorphism was higher in diabetic patients compared to controls (CT: P < 0.0001, OR = 3.01; TT: P < 0.0001, OR = 2.45). IL-1β (C-511T) polymorphism was linked with plasma IL-1β levels. Interestingly, heterozygous mutants (CT) were most prevalent in T2D individuals with CAD compared to those without CAD ( P = 0.03, OR = 1.82). Furthermore, low-density lipoprotein (LDL) and triglycerides were elevated in T2D patients with CAD than in patients without heart-related disorder. No significant association of other polymorphism (T-31C) was noticed with susceptibility to T2D or diabetic patients with heart disorders. IL-1β (C-511T) variants are associated with elevated plasma IL-1β levels. Mutation at the IL-1β promoter region (C-511T) predisposed subjects to the development of T2D and CAD manifestation in diabetic patients.
Collapse
Affiliation(s)
- Lijing Ma
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan, China
- Department of Endocrinology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Heng Su
- Department of Endocrinology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Yan Wang
- Department of Endocrinology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Yikun Zhou
- Department of Endocrinology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Zhuang Kang
- Department of Endocrinology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Ying Xu
- Department of Endocrinology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Jie Gao
- Department of Endocrinology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| |
Collapse
|
38
|
Halenova T, Zlatskiy I, Syroeshkin A, Maximova T, Pleteneva T. Deuterium-Depleted Water as Adjuvant Therapeutic Agent for Treatment of Diet-Induced Obesity in Rats. Molecules 2019; 25:E23. [PMID: 31861678 PMCID: PMC6982901 DOI: 10.3390/molecules25010023] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/03/2019] [Accepted: 12/12/2019] [Indexed: 12/26/2022] Open
Abstract
In this study, we present the potential application of deuterium-depleted water (DDW) for the prevention and adjuvant treatment of obesity in rats. We tested the hypothesis that DDW can alleviate diet-induced obesity (DIO) and its associated metabolic impairments. Rats fed a high-fat diet had an increased body weight index (BWI), glucose concentration, and level of certain proinflammatory cytokines; decreased levels of insulin in the serum; decreased tryptophan and serotonin in the brain, and a decreased concentration of some heavy metals in the liver. Drinking DDW at a concentration of 10 ppm deuterium/protium (D/H) ad libitum for 3 weeks restored the BWI, glucose (serum), tryptophan (brain), and serotonin (brain) levels and concentration of Zn in the liver in the DIO animals to those of the controls. The levels of proinflammatory cytokines (IL-1β, IL-6, IFNγ) and anti-inflammatory TNFα were decreased in DIO rats, while anti-inflammatory cytokine (IL-4, IL-10) levels remained at the control levels, which is indicative of a pathophysiological syndrome. In contrast, in groups of rats treated with DDW, a significant increase in anti-inflammatory (IL-4, IL-10) and proinflammatory cytokines (IFNγ) was observed. This finding indicates a reduction in systemic inflammation in obese animals treated with DDW. Similarly, the high-fat diet caused an increased level of oxidative stress products, which was accompanied by decreased activity of both superoxide dismutase and catalase, whereas the administration of DDW decreased the level of oxidative stress and enhanced antioxidant enzyme activities.
Collapse
Affiliation(s)
- Tetiana Halenova
- Taras Shevchenko National University of Kyiv, ESC “Institute of Biology and Medicine”, 64 Volodymyrska Str., 01601 Kyiv, Ukraine;
| | - Igor Zlatskiy
- Peoples Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya St, Moscow 117198, Russian; (A.S.); (T.M.); (T.P.)
- State Institute of Genetic and Regenerative Medicine NAMS of Ukraine, 67 Vyshgorodska Str., 04114 Kyiv, Ukraine
| | - Anton Syroeshkin
- Peoples Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya St, Moscow 117198, Russian; (A.S.); (T.M.); (T.P.)
| | - Tatiana Maximova
- Peoples Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya St, Moscow 117198, Russian; (A.S.); (T.M.); (T.P.)
| | - Tatiana Pleteneva
- Peoples Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya St, Moscow 117198, Russian; (A.S.); (T.M.); (T.P.)
| |
Collapse
|
39
|
Đorđević M, Grdović N, Mihailović M, Arambašić Jovanović J, Uskoković A, Rajić J, Sinadinović M, Tolić A, Mišić D, Šiler B, Poznanović G, Vidaković M, Dinić S. Centaurium erythraea extract improves survival and functionality of pancreatic beta-cells in diabetes through multiple routes of action. JOURNAL OF ETHNOPHARMACOLOGY 2019; 242:112043. [PMID: 31252092 DOI: 10.1016/j.jep.2019.112043] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/24/2019] [Accepted: 06/24/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Centaurium erythraea Rafn (CE) is used as a traditional medicinal plant in Serbia to treat different ailments due to its antidiabetic, antipyretic, antiflatulent and detoxification effects. AIM OF THE STUDY Elucidation of the mechanisms that underlie the antioxidant and pro-survival effects of the CE extract (CEE) in beta-cells and pancreatic islets from streptozotocin (STZ)-treated diabetic rats. MATERIAL AND METHODS Diabetes was induced in rats by multiple applications of low doses of STZ (40 mg/kg intraperitoneally (i.p.), for five consecutive days). CEE (100 mg/kg) was administered orally, in the pre-treated group for two weeks before diabetes induction, during the treatments with STZ and for four weeks after diabetes onset, and in the post-treatment group for four weeks after diabetes induction. The impact of CEE on diabetic islets was estimated by histological and immunohistochemical examination of the pancreas. Molecular mechanisms of the effects of CEE were also analyzed in insulinoma Rin-5F cells treated with STZ (12 mM) and CEE (0.25 mg/mL). Oxidative stress was evaluated by assessing the levels of DNA damage, lipid peroxidation, protein S-glutathionylation and enzymatic activities and expression of CAT, MnSOD, CuZnSOD, GPx and GR in beta-cells. The presence and activities of the redox-sensitive and islet-enriched regulatory proteins were also analyzed. RESULTS Treatment with CEE ameliorated the insulin level and glycemic control in STZ-induced diabetic rats by improving the structural and functional properties of pancreatic islets through multiple routes of action. The disturbance of islet morphology and islet cell contents in diabetes was reduced by the CEE treatment and was associated with a protective effect of CEE on the levels of insulin, GLUT-2 and p-Akt in diabetic islets. The antioxidant effect of CEE on STZ-treated beta-cells was displayed as reduced DNA damage, lipid peroxidation, protein S-glutathionylation and alleviation of STZ-induced disruption in MnSOD, CuZnSOD and CAT enzyme activities. The oxidative stress-induced disturbance of the transcriptional regulation of CAT, MnSOD, CuZnSOD, GPx and GR enzymes in beta-cells was improved after the CEE treatment, and was observed as readjustment of the presence and activities of redox-sensitive NFκB-p65, FOXO3A, Sp1 and Nrf-2 transcription factors. The observed CEE-mediated induction of proliferative and pro-survival pathways and insulin expression/secretion after STZ-induced oxidative stress in beta-cells could be partially attributed to a fine-tuned modulation of the activities of pro-survival Akt, ERK and p38 kinases and islet-enriched Pdx-1 and MafA regulatory factors. CONCLUSIONS The results of this study provide evidence that CEE improves the structural and functional properties of pancreatic beta-cells by correcting the endogenous antioxidant regulatory mechanisms and by promoting proliferative and pro-survival pathways in beta-cells.
Collapse
Affiliation(s)
- Miloš Đorđević
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia.
| | - Nevena Grdović
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia.
| | - Mirjana Mihailović
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia.
| | - Jelena Arambašić Jovanović
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia.
| | - Aleksandra Uskoković
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia.
| | - Jovana Rajić
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia.
| | - Marija Sinadinović
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia.
| | - Anja Tolić
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia.
| | - Danijela Mišić
- Department of Plant Physiology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia.
| | - Branislav Šiler
- Department of Plant Physiology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia.
| | - Goran Poznanović
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia.
| | - Melita Vidaković
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia.
| | - Svetlana Dinić
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia.
| |
Collapse
|
40
|
Donath MY, Dinarello CA, Mandrup-Poulsen T. Targeting innate immune mediators in type 1 and type 2 diabetes. Nat Rev Immunol 2019; 19:734-746. [PMID: 31501536 DOI: 10.1038/s41577-019-0213-9] [Citation(s) in RCA: 204] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2019] [Indexed: 02/07/2023]
Abstract
Type 1 and type 2 diabetes are characterized by chronic inflammation; both diseases involve pancreatic islet inflammation, while systemic low-grade inflammation is a feature of obesity and type 2 diabetes. Long-term activation of the innate immune system impairs insulin secretion and action, and inflammation also contributes to macrovascular and microvascular complications of diabetes. However, despite strong preclinical evidence and proof-of-principle clinical trials demonstrating that targeting inflammatory pathways can prevent cardiovascular disease and other complications in patients with diabetes, there are still no approved treatments for diabetes that target innate immune mediators. Here, we review recent advances in our understanding of the inflammatory pathogenesis of type 1 and type 2 diabetes from a translational angle and point out the critical gaps in knowledge that need to be addressed to guide drug development.
Collapse
Affiliation(s)
- Marc Y Donath
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland. .,Department of Biomedicine, University of Basel, Basel, Switzerland.
| | | | | |
Collapse
|
41
|
Tamarai K, Bhatti JS, Reddy PH. Molecular and cellular bases of diabetes: Focus on type 2 diabetes mouse model-TallyHo. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2276-2284. [PMID: 31082469 DOI: 10.1016/j.bbadis.2019.05.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 12/17/2022]
Abstract
Diabetes is a chronic lifestyle disorder that affects millions of people worldwide. Diabetes is a condition where the body does not produce sufficient insulin or does not use it efficiently. Insulin resistance in diabetes or obesity causes the pancreatic β-cells to increase the insulin output. Diabetes occurs in multiple forms, including type 1, type 2, type 3 and gestational. Type 2 diabetes accounts for ∼90-95% of total affected population and is associated with both impaired insulin production by the β-cells of the pancreas and impaired insulin release in response to high blood glucose levels. Diabetes is tightly linked with genetic mutations and genetic and lifestyle activities, including diet and exercise. Recent epidemiological studies established a close link between the diabetes and progression to Alzheimer's disease. This article summarizes various molecular mechanisms involved in the developments of diabetes, including biochemical characteristics, genetic and molecular links with Alzheimer's disease, β-cell function, and factors associated with diabetes. This will help us in the development of novel therapeutic strategies targeting AD in future.
Collapse
Affiliation(s)
- Kavya Tamarai
- Internal Medicine Department, Texas Tech University Health Sciences Center, 3601 4(th) Street, Lubbock, TX 79430, United States
| | - Jasvinder Singh Bhatti
- Internal Medicine Department, Texas Tech University Health Sciences Center, 3601 4(th) Street, Lubbock, TX 79430, United States; Department of Biotechnology, Sri Guru Gobind Singh College, Chandigarh, India
| | - P Hemachandra Reddy
- Internal Medicine Department, Texas Tech University Health Sciences Center, 3601 4(th) Street, Lubbock, TX 79430, United States; Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States; Cell Biology & Biochemistry Department, Texas Tech University Health Sciences Center, 3601 4(th) Street, Lubbock, TX 79430, United States; Pharmacology & Neuroscience Department, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States; Neurology Department, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States; Speech, Language and Hearing Sciences Department, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States; Department of Public Health, Graduate School of Biomedical Sciences, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States.
| |
Collapse
|
42
|
Turpaev K, Krizhanovskii C, Wang X, Sargsyan E, Bergsten P, Welsh N. The protein synthesis inhibitor brusatol normalizes high-fat diet-induced glucose intolerance in male C57BL/6 mice: role of translation factor eIF5A hypusination. FASEB J 2019; 33:3510-3522. [PMID: 30462531 DOI: 10.1096/fj.201801698r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The naturally occurring quassinoid compound brusatol improves the survival of insulin-producing cells when exposed to the proinflammatory cytokines IL-1β and IFN-γ in vitro. The aim of the present study was to investigate whether brusatol also promotes beneficial effects in mice fed a high-fat diet (HFD), and if so, to study the mechanisms by which brusatol acts. In vivo, we observed that the impaired glucose tolerance of HFD-fed male C57BL/6 mice was counteracted by a 2 wk treatment with brusatol. Brusatol treatment improved both β-cell function and peripheral insulin sensitivity of HFD-fed mice. In vitro, brusatol inhibited β-cell total protein and proinsulin biosynthesis, with an ED50 of ∼40 nM. In line with this, brusatol blocked cytokine-induced iNOS protein expression via inhibition of iNOS mRNA translation. Brusatol may have affected protein synthesis, at least in part, via inhibition of eukaryotic initiation factor 5A (eIF5A) hypusination, as eIF5A spermidine association and hypusination in RIN-5AH cells was reduced in a dose- and time-dependent manner. The eIF5A hypusination inhibitor GC7 promoted a similar effect. Both brusatol and GC7 protected rat RIN-5AH cells against cytokine-induced cell death. Brusatol reduced eIF5A hypusination and cytokine-induced cell death in EndoC-βH1 cells as well. Finally, hypusinated eIF5A was reduced in vivo by brusatol in islet endocrine and endothelial islet cells of mice fed an HFD. The results of the present study suggest that brusatol improves glucose intolerance in mice fed an HFD, possibly by inhibiting protein biosynthesis and eIF5A hypusination.-Turpaev, K., Krizhanovskii, C., Wang, X., Sargsyan, E., Bergsten, P., Welsh, N. The protein synthesis inhibitor brusatol normalizes high-fat diet-induced glucose intolerance in male C57BL/6 mice: role of translation factor eIF5A hypusination.
Collapse
Affiliation(s)
- Kyril Turpaev
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden; and
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, Russia
| | - Camilla Krizhanovskii
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden; and
| | - Xuan Wang
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden; and
| | - Ernest Sargsyan
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden; and
| | - Peter Bergsten
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden; and
| | - Nils Welsh
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden; and
| |
Collapse
|
43
|
Ying W, Lee YS, Dong Y, Seidman JS, Yang M, Isaac R, Seo JB, Yang BH, Wollam J, Riopel M, McNelis J, Glass CK, Olefsky JM, Fu W. Expansion of Islet-Resident Macrophages Leads to Inflammation Affecting β Cell Proliferation and Function in Obesity. Cell Metab 2019; 29:457-474.e5. [PMID: 30595478 PMCID: PMC6701710 DOI: 10.1016/j.cmet.2018.12.003] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 09/27/2018] [Accepted: 11/30/2018] [Indexed: 02/07/2023]
Abstract
The nature of obesity-associated islet inflammation and its impact on β cell abnormalities remains poorly defined. Here, we explore immune cell components of islet inflammation and define their roles in regulating β cell function and proliferation. Islet inflammation in obese mice is dominated by macrophages. We identify two islet-resident macrophage populations, characterized by their anatomical distributions, distinct phenotypes, and functional properties. Obesity induces the local expansion of resident intra-islet macrophages, independent of recruitment from circulating monocytes. Functionally, intra-islet macrophages impair β cell function in a cell-cell contact-dependent manner. Increased engulfment of β cell insulin secretory granules by intra-islet macrophages in obese mice may contribute to restricting insulin secretion. In contrast, both intra- and peri-islet macrophage populations from obese mice promote β cell proliferation in a PDGFR signaling-dependent manner. Together, these data define distinct roles and mechanisms for islet macrophages in the regulation of islet β cells.
Collapse
Affiliation(s)
- Wei Ying
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Yun Sok Lee
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Yi Dong
- Pediatric Diabetes Research Center, Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Jason S Seidman
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Meixiang Yang
- Pediatric Diabetes Research Center, Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; The First Affiliated Hospital, Biomedical Translational Research Institute, Jinan University, Guangzhou 510632, China
| | - Roi Isaac
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Jong Bae Seo
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Bi-Huei Yang
- Pediatric Diabetes Research Center, Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Joshua Wollam
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Matthew Riopel
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Joanne McNelis
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Christopher K Glass
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Jerrold M Olefsky
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| | - Wenxian Fu
- Pediatric Diabetes Research Center, Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
44
|
Lee HA, Lee E, Do GY, Moon EK, Quan FS, Kim I. Histone deacetylase inhibitor MGCD0103 protects the pancreas from streptozotocin-induced oxidative stress and β-cell death. Biomed Pharmacother 2018; 109:921-929. [PMID: 30551546 DOI: 10.1016/j.biopha.2018.10.163] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/22/2018] [Accepted: 10/25/2018] [Indexed: 12/13/2022] Open
Abstract
Inhibition of histone deacetylase (HDAC) suppresses inflammation of pancreatic islets and apoptosis of β-cells. However, the underlying molecular mechanism is unclear. In the present study, we demonstrate that MGCD0103 (MGCD), an HDAC inhibitor, protects the pancreas from streptozotocin (STZ)-induced oxidative stress and cell death. Sprague-Dawley rats were intraperitoneally injected with STZ (40 mg/kg) to induce type I diabetes. MGCD (10 μg/day) was infused with osmotic mini-pump for 4 weeks. Pancreatic insulin and macrophage infiltration were analyzed by immunohistochemistry. Cellular level of reactive oxygen species (ROS) was evaluated with fluorescence-activated cell sorting. Tetramethylrhodamine ethyl ester was used to analyze mitochondrial membrane potential. Activation of caspase-3 was analyzed by western blotting. Chromatin immunoprecipitation was performed to investigate the binding affinity of specificity protein 1 (SP1) on the promoters of target genes. mRNA expression was analyzed by quantitative real-time polymerase chain reaction. As a result, we found that MGCD infusion ameliorated STZ-induced hyperglycemia, islet deformation, decreased insulin level, and macrophage infiltration. STZ injection promoted the production of ROS, which induced caspase activity and β-cell death. 4-Hydroxy-2,2,6,6-tetramethylpiperidin-1-oxyl (TEMPOL), a mimetic of superoxide dismutase (SOD), reduced STZ-induced caspase activity and β-cell death. MGCD treatment increased SOD expression and histone acetylation level on promoters. Infusion of MGCD promoted acetylation of SP1 and its enrichment on SOD promoters. Thus, MGCD protects pancreatic β-cells from STZ-induced oxidative stress and cell death through the induction of antioxidant enzymes such as SODs.
Collapse
Affiliation(s)
- Hae-Ahm Lee
- Department of Pharmacology, Cardiovascular Research Institute, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Eunjo Lee
- Department of Pharmacology, Cardiovascular Research Institute, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Ga Young Do
- Department of Pharmacology, Cardiovascular Research Institute, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Eun-Kyung Moon
- Department of Medical Zoology, Kyung Hee University School of Medicine, Seoul 02447, Republic of Korea
| | - Fu-Shi Quan
- Department of Medical Zoology, Kyung Hee University School of Medicine, Seoul 02447, Republic of Korea
| | - Inkyeom Kim
- Department of Pharmacology, Cardiovascular Research Institute, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea.
| |
Collapse
|
45
|
Shi S, Zhao L, Zheng L. NSD2 is downregulated in T2DM and promotes β cell proliferation and insulin secretion through the transcriptionally regulation of PDX1. Mol Med Rep 2018; 18:3513-3520. [PMID: 30066931 DOI: 10.3892/mmr.2018.9338] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 06/27/2018] [Indexed: 11/06/2022] Open
Abstract
Diabetes has become a major public health issue in the world. Type 2 diabetes mellitus (T2DM), also known as non‑insulin‑dependent diabetes mellitus, has been identified to result in an inability to compensate for insulin resistance. A previous study has shown that NSD2 regulates glucose metabolism; however, whether NSD2 serves roles in diabetes has not been thoroughly elucidated to date. In present study, the expression of NSD2 in blood samples from patients with T2DM was compared with that in healthy volunteers. Notably, the expression of NSD2 was negatively correlated with glucose concentration but positively associated with PDX1 expression. Several functional experiments, including CCK‑8 assay and colony formation assay, revealed that NSD2 promoted the proliferation of pancreatic β cell lines. Moreover, ectopic expression of NSD2 significantly promoted insulin secretion. In addition, NSD2 served as a transfection factor and it was identified that NSD2 transcriptionally regulated PDX1 expression through its H3K36me2 methyltransferase activity. The present study indicated that NSD2 may be a novel molecular therapy target of T2DM.
Collapse
Affiliation(s)
- Suqin Shi
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Lu Zhao
- Department of Endocrinology, The Third Affiliated Hospital of Henan University of TCM, Zhengzhou, Henan 450004, P.R. China
| | - Lili Zheng
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
46
|
Kim DS, Song L, Wang J, Wu H, Gou W, Cui W, Kim JS, Wang H. Carbon Monoxide Inhibits Islet Apoptosis via Induction of Autophagy. Antioxid Redox Signal 2018; 28:1309-1322. [PMID: 28826228 PMCID: PMC5905947 DOI: 10.1089/ars.2016.6979] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
AIM Carbon monoxide (CO) functions as a therapeutic molecule in various disease models because of its anti-inflammatory and antiapoptotic properties. We investigated the capacity of CO to reduce hypoxia-induced islet cell death and dysfunction in human and mouse models. RESULTS Culturing islets in CO-saturated medium protected them from hypoxia-induced apoptosis and preserved β cell function by suppressing expression of proapoptotic (Bim, PARP, Cas-3), proinflammatory (TNF-α), and endoplasmic reticulum (ER) stress (glucose-regulated protein 94, grp94, CHOP) proteins. The prosurvival effects of CO on islets were attenuated when autophagy was blocked by specific inhibitors or when either ATG7 or ATG16L1, two essential factors for autophagy, was downregulated by siRNA. In vivo, CO exposure reduced both inflammation and cell death in grafts immediately after transplantation, and enhanced long-term graft survival of CO-treated human and mouse islet grafts in streptozotocin-induced diabetic non-obese diabetic severe combined immunodeficiency (NOD-SCID) or C57BL/6 recipients. INNOVATION These findings underline that pretreatment with CO protects islets from hypoxia and stress-induced cell death via upregulation of ATG16L1-mediated autophagy. CONCLUSION Our results suggested that CO exposure may provide an effective means to enhance survival of grafts in clinical islet cell transplantation, and may be beneficial in other diseases in which inflammation and cell death pose impediments to achieving optimal therapeutic effects. Antioxid. Redox Signal. 28, 1309-1322.
Collapse
Affiliation(s)
- Do-Sung Kim
- 1 Department of Surgery, Medical University of South Carolina , Charleston, South Carolina
| | - Lili Song
- 1 Department of Surgery, Medical University of South Carolina , Charleston, South Carolina
| | - Jingjing Wang
- 1 Department of Surgery, Medical University of South Carolina , Charleston, South Carolina
| | - Hongju Wu
- 2 Department of Medicine, Tulane University , New Orleans, Louisiana
| | - Wenyu Gou
- 1 Department of Surgery, Medical University of South Carolina , Charleston, South Carolina
| | - Wanxing Cui
- 3 Medstar Georgetown University Hospital , Washington DC
| | - Jae-Sung Kim
- 4 Department of Surgery, University of Florida , Gainesville, Florida
| | - Hongjun Wang
- 1 Department of Surgery, Medical University of South Carolina , Charleston, South Carolina
| |
Collapse
|
47
|
Effect of C 60 fullerene nanoparticles on the diet-induced obesity in rats. Int J Obes (Lond) 2018; 42:1987-1998. [PMID: 30401827 DOI: 10.1038/s41366-018-0016-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 11/13/2017] [Accepted: 12/27/2017] [Indexed: 01/04/2023]
Abstract
BACKGROUND Obesity is a growing global health problem. Since increased oxidative stress is one of the key pathological mechanisms underpinning overweight and strongly correlates with progression of obesity-related complications we hypothesized that C60 fullerene nanoparticles, due to their strong antioxidant capacity, could be the promising therapeutic agent in the treatment of this disease. Here we investigated whether the C60 fullerenes can alleviate diet-induced obesity (DIO) and metabolic impairments associated with it. METHODS To determine the effect of C60 fullerenes on some nutritional and metabolic parameters, rats were fed either a normal diet (6.7% fat, 15.27 kJ·g-1) or a high-fat diet (38.8% fat, 28.71 kJ·g-1) for 70 days and were simultaneously treated per os with pristine C60 fullerene aqueous solution (C60FAS; 0.3 mg·kg-1 every other day) since the 28th day from the start of the experiment. RESULTS Rats fed with high fat diet had significantly increased body mass index (BMI), levels of insulin, glucose, glycosilated hemoglobin (HbA1c) and serum pro-inflammatory cytokines compared with control rats fed with low-fat chow. C60 fullerenes normalized the metabolic parameters and partially reduced BMI in DIO animals. Pro-inflammatory cytokines (IL-1b, IL-12, INFγ) were also decreased in serum of DIO rats treated with C60 fullerenes while anti-inflammatory cytokines (IL-4, IL-10) were at the control levels. High fat diet caused the increased level of oxidative stress products, and this was accompanied by decreased activity both the superoxide dismutase and catalase, whereas the administration of C60 fullerenes markedly decreased level of oxidative stress and enhanced antioxidant enzyme activities. CONCLUSION These data indicate that water-soluble pristine C60 fullerenes reduce chronic inflammation, restore glucose homeostasis as well as positively affects on prooxidant-antioxidant homeostasis. C60 fullerenes could be represented as a promising therapeutic agent in the treatment of obesity and its related complications.
Collapse
|
48
|
Bogdani M, Blackman SM, Ridaura C, Bellocq JP, Powers AC, Aguilar-Bryan L. Structural abnormalities in islets from very young children with cystic fibrosis may contribute to cystic fibrosis-related diabetes. Sci Rep 2017; 7:17231. [PMID: 29222447 PMCID: PMC5722914 DOI: 10.1038/s41598-017-17404-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 11/22/2017] [Indexed: 01/18/2023] Open
Abstract
Cystic fibrosis (CF)-related diabetes (CFRD) is thought to result from beta-cell injury due in part to pancreas exocrine damage and lipofibrosis. CFRD pancreata exhibit reduced islet density and altered cellular composition. To investigate a possible etiology, we tested the hypothesis that such changes are present in CF pancreata before the development of lipofibrosis. We evaluated pancreas and islet morphology in tissues from very young CF children (<4 years of age), and adult patients with CF and CFRD. The relative number of beta-cells in young CF tissues was reduced by 50% or more when compared to age-matched controls. Furthermore, young CF tissues displayed significantly smaller insulin-positive areas, lower proportion of beta-cells positive for the proliferation marker Ki67 or the ductal marker CK19 vs. control subjects, and islet inflammatory cell infiltrates, independently of the severity of the exocrine lesion and in the absence of amyloid deposits. CFRD pancreata exhibited greater islet injury with further reduction in islet density, decreased relative beta-cell number, and presence of amyloid deposits. Together, these results strongly suggest that an early deficiency in beta-cell number in infants with CF may contribute to the development of glucose intolerance in the CF pediatric population, and to CFRD, later in life.
Collapse
Affiliation(s)
| | - Scott M Blackman
- Division of Pediatric Endocrinology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cecilia Ridaura
- Department of Pathology, Instituto Nacional de Pediatría, Mexico City, Mexico
| | | | - Alvin C Powers
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, and Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, and VA Tennessee Valley Healthcare System, Nashville, TN, USA
| | | |
Collapse
|
49
|
Søgaard KL, Ellervik C, Svensson J, Thorsen SU. The Role of Iron in Type 1 Diabetes Etiology: A Systematic Review of New Evidence on a Long-Standing Mystery. Rev Diabet Stud 2017; 14:269-278. [PMID: 29145537 PMCID: PMC6115007 DOI: 10.1900/rds.2017.14.269] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/13/2017] [Accepted: 08/24/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The incidence of type 1 diabetes (T1D) is rising, which might be due to the influence of environmental factors. Biological and epidemiological evidence has shown that excess iron is associated with beta-cell damage and impaired insulin secretion. AIM In this review, our aim was to assess the association between iron and the risk of T1D. METHODS A systematic literature search was performed in PubMed and EMBASE in July 2016. Studies investigating the effect of iron status/intake on the risk of developing T1D later were included, and study quality was evaluated. The results have been summarized in narrative form. RESULTS From a total of 931 studies screened, we included 4 observational studies evaluating iron intake from drinking water or food during early life and the risk of T1D. The quality of the studies was moderate to high assessed via the nine-star Newcastle Ottawa Scale. One out of the four studies included in this review found estimates of dietary iron intake to be associated with risk of T1D development, whereas three studies found no such relationship for estimates of iron in drinking water. CONCLUSIONS The limited number of studies included found dietary iron, but not iron in drinking water, to be associated with risk of T1D. Further studies are needed to clarify the association between iron and risk of T1D, especially studies including measurements of body iron status.
Collapse
Affiliation(s)
- Karen L. Søgaard
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - Christina Ellervik
- Department of Production, Research, and Innovation; Region Zealand, Alleen 15, 4180 Sorø, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
- Department of Laboratory Medicine, Boston Children`s Hospital, 300 Longwood Avenue, 02115, Boston, MA, USA
- Harvard Medical School, 25 Shattuck St, 02115, Boston, MA, USA
| | - Jannet Svensson
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Steffen U. Thorsen
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730 Herlev, Denmark
| |
Collapse
|
50
|
El-Shemi AG, Kensara OA, Alsaegh A, Mukhtar MH. Pharmacotherapy with Thymoquinone Improved Pancreatic β-Cell Integrity and Functional Activity, Enhanced Islets Revascularization, and Alleviated Metabolic and Hepato-Renal Disturbances in Streptozotocin-Induced Diabetes in Rats. Pharmacology 2017; 101:9-21. [DOI: 10.1159/000480018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 07/31/2017] [Indexed: 12/20/2022]
Abstract
Aims: This study is aimed at evaluating the antidiabetic effects of thymoquinone (TQ) on streptozotocin (STZ)-induced diabetes in rats, and exploring the possible underlying mechanisms. Methods: Diabetes was induced in adult male Wistar rats by intraperitoneal injection of freshly prepared STZ (65 mg/kg). After disease induction, 42 rats were equally assigned to: controls, STZ-diabetic group, and STZ-diabetic group treated with oral TQ (35 mg/kg/day) for 5 weeks. Fasting blood glucose levels were determined weekly, and the animals were euthanized at day 38 post-STZ injection. Blood samples were assessed for glucose-insulin homeostasis parameters (plasma glucose, glycated hemoglobin, serum insulin, homeostatic model assessment of insulin resistance, and insulin sensitivity index) and lipid profile. Resected pancreases were subjected to histological examination and immunohistochemical or enzyme-linked immunosorbent assay assessment to determine the pancreatic expression of insulin sensitizing β-cells, anti-apoptotic protein “survivin,” apoptosis-inducer “caspase-3,” prototypic angiogenic factors (vascular endothelial growth factor [VEGF] and endothelial cluster of differentiation 31 [CD31]), pro- and anti-inflammatory cytokines (interleukin-1beta [IL-1β] and interleukin-10 [IL-10], respectively), thiobarbituric acid reactive substances (TBARS), total glutathione (GSH), and superoxide dismutase (SOD). The hepato-renal statuses were assessed biochemically and histologically. Results: Therapy with TQ markedly improved the integrity of pancreatic islets, glucose-insulin homeostasis-related parameters, lipid profile parameters, and hepato-renal functional and histomorphological statuses that collectively were severely deteriorated in untreated diabetic group. Mechanistically, TQ therapy efficiently increased insulin producing β-cells, upregulated survivin, VEGF, CD31, IL-10, GSH and SOD, and downregulated caspase-3, IL-1β, and TBARSs in the pancreatic tissues of STZ-diabetic rats. Conclusions: These findings prove the anti-diabetic potential of TQ and its efficacy in regenerating pancreatic β-cells and ameliorating pancreatic inflammation and oxidative stress, and highlight its novelty in repressing apoptosis of β-cells and enhancing islet revascularization in STZ-diabetic rats. Further studies are required to support these findings and realize their possible clinical significance.
Collapse
|