1
|
Bhowmick DC, Ahn M, Bhattacharya S, Aslamy A, Thurmond DC. DOC2b enrichment mitigates proinflammatory cytokine-induced CXCL10 expression by attenuating IKKβ and STAT-1 signaling in human islets. Metabolism 2025; 164:156132. [PMID: 39805534 PMCID: PMC11798586 DOI: 10.1016/j.metabol.2025.156132] [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: 08/06/2024] [Revised: 12/22/2024] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
INTRODUCTION Type 1 diabetic human islet β-cells are deficient in double C 2 like domain beta (DOC2b) protein. Further, DOC2b protects against cytokine-induced pancreatic islet β-cell stress and apoptosis. However, the mechanisms underpinning the protective effects of DOC2b remain unknown. METHODS Biochemical studies, qPCR, proteomics, and immuno-confocal microscopy were conducted to determine the underlying protective mechanisms of DOC2b in β-cells. DOC2b-enriched or -depleted primary islets (human and mouse) and β-cell lines challenged with or without proinflammatory cytokines, global DOC2b heterozygous knockout mice subjected to multiple-low-dose-streptozotocin (MLD-STZ), were used for these studies. RESULTS A significant elevation of stress-induced CXCL10 mRNA was observed in DOC2b-depleted β-cells and primary mouse islets. Further, DOC2b enrichment markedly attenuated cytokine-induced CXCL10 levels in primary non-diabetic human islets and β-cells. DOC2b enrichment also reduced total-NF-κB p65 protein levels in human islets challenged with T1D mimicking proinflammatory cytokines. IKKβ, NF-κB p65, and STAT-1 are capable of associating with DOC2b in cytokine-challenged β-cells. DOC2b enrichment in cytokine-stressed human islets and β-cells corresponded with a significant reduction in activated and total IKKβ protein levels. Total IκBβ protein was increased in DOC2b-enriched human islets subjected to acute cytokine challenge. Cytokine-induced activated and total STAT-1 protein and mRNA levels were markedly reduced in DOC2b-enriched human islets. Intriguingly, DOC2b also prevents ER-stress-IKKβ and STAT-1 crosstalk in the rat INS1-832/13 β-cell line. CONCLUSION The mechanisms underpinning the protective effects of DOC2b involve attenuation of IKKβ-NF-κB p65 and STAT-1 signaling, and reduced CXCL10 expression.
Collapse
Affiliation(s)
- Diti Chatterjee Bhowmick
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA, USA.
| | - Miwon Ahn
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Supriyo Bhattacharya
- Shared Resources-Integrative Genomics, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Arianne Aslamy
- Department of Medicine, Cedars-Sinai Medical Center, West Hollywood, CA, USA
| | - Debbie C Thurmond
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA, USA.
| |
Collapse
|
2
|
Perez-Serna AA, Guzman-Llorens D, Dos Santos RS, Marroqui L. Bcl-2 and Bcl-xL in Diabetes: Contributions to Endocrine Pancreas Viability and Function. Biomedicines 2025; 13:223. [PMID: 39857806 PMCID: PMC11760435 DOI: 10.3390/biomedicines13010223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/08/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Diabetes is a chronic metabolic disorder whose prevalence increases every year, affecting more than 530 million adults worldwide. Type 1 (T1D) and type 2 diabetes (T2D), the most common forms of diabetes, are characterized by the loss of functional pancreatic β-cells, mostly due to apoptosis. B-cell leukemia/lymphoma 2 (Bcl-2) and B-cell lymphoma-extra large (Bcl-xL), two anti-apoptotic proteins belonging to the Bcl-2 family, are crucial for regulating the intrinsic pathway of apoptosis. However, over the years, they have been implicated in many other cellular processes, including intracellular Ca2+ homeostasis and the regulation of mitochondrial metabolism. Thus, understanding the biological processes in which these proteins are involved may be crucial to designing new therapeutic targets. This review summarizes the roles of Bcl-2 and Bcl-xL in apoptosis and metabolic homeostasis. It focuses on how the dysregulation of Bcl-2 and Bcl-xL affects pancreatic β-cell function and survival, and the consequences for diabetes development.
Collapse
Affiliation(s)
- Atenea A. Perez-Serna
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain; (A.A.P.-S.); (D.G.-L.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | - Daniel Guzman-Llorens
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain; (A.A.P.-S.); (D.G.-L.)
| | - Reinaldo S. Dos Santos
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain; (A.A.P.-S.); (D.G.-L.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l’Almazara 11, 03203 Elche, Alicante, Spain
| | - Laura Marroqui
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain; (A.A.P.-S.); (D.G.-L.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
3
|
Duan F, Wu J, Chang J, Peng H, Liu Z, Liu P, Han X, Sun T, Shang D, Yang Y, Li Z, Li P, Liu Y, Zhu Y, Lv Y, Guo X, Zhao Y, An Y. Deciphering endocrine function of adipose tissue and its significant influences in obesity-related diseases caused by its dysfunction. Differentiation 2025; 141:100832. [PMID: 39709882 DOI: 10.1016/j.diff.2024.100832] [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: 10/29/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024]
Abstract
Current research has found that adipose tissue is not only involved in energy metabolism, but also a highly active endocrine organ that secretes various adipokines, including adiponectin, leptin, resistin and apelin, which are involved in the regulation of physiology and pathology of tissues and organs throughout the body. With the yearly increasing incidence, obesity has become a risk factor for a variety of pathological changes, including inflammation and metabolic syndrome in various system (endocrine, circulatory, locomotor and central nervous system). Thus these symptoms lead to multi-organ dysfunctions, including the heart, liver, kidneys, brain and joints. An in-depth summary of the roles of adipokines in the regulation of other tissues and organs can help to provide more effective therapeutic strategies for obesity-related diseases and explore potential therapeutic targets. Therefore, this review has retrospected the endocrine function of adipose tissue under obesity and the role of dysregulated adipokine secretion in related diseases and the underlying mechanisms, in order to provide a theoretical basis for targeting adipokine-mediated systemic dysregulation.
Collapse
Affiliation(s)
- Feiyi Duan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Jiaoyan Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Jiayi Chang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Haoyuan Peng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Zitao Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Pengfei Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Xu Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Tiantian Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Dandan Shang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Yutian Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Zhihao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Pengkun Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Yixuan Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Yonghao Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Yunzhi Lv
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Xiumei Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Ying Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
4
|
Chatterjee Bhowmick D, Ahn M, Bhattacharya S, Aslamy A, Thurmond DC. DOC2b enrichment mitigates proinflammatory cytokine-induced CXCL10 expression by attenuating IKKβ and STAT-1 signaling in human islets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.22.629540. [PMID: 39763877 PMCID: PMC11703217 DOI: 10.1101/2024.12.22.629540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Introduction Type 1 diabetic human islet β-cells are deficient in double C 2 like domain beta (DOC2b) protein. Further, DOC2b protects against cytokine-induced pancreatic islet β-cell stress and apoptosis. However, the mechanisms underpinning the protective effects of DOC2b remain unknown. Methods Biochemical studies, qPCR, proteomics, and immuno-confocal microscopy were conducted to determine the underlying protective mechanisms of DOC2b in β-cells. DOC2b- enriched or-depleted primary islets (human and mouse) and β-cell lines challenged with or without proinflammatory cytokines, global DOC2b heterozygous knockout mice subjected to multiple-low-dose-streptozotocin (MLD-STZ), were used for these studies. Results A significant elevation of stress-induced CXCL10 mRNA was observed in DOC2b- depleted β-cells and primary mouse islets. Further, DOC2b enrichment markedly attenuated cytokine-induced CXCL10 levels in primary non-diabetic human islets and β-cells. DOC2b enrichment also reduced total-NF-κB p65 protein levels in human islets challenged with T1D mimicking proinflammatory cytokines. IKKβ, NF-κB p65, and STAT-1 are capable of associating with DOC2b in cytokine-challenged β-cells. DOC2b enrichment in cytokine-stressed human islets and β-cells corresponded with a significant reduction in activated and total IKKβ protein levels. Total IκBβ protein was increased in DOC2b-enriched human islets subjected to acute cytokine challenge. Cytokine-induced activated and total STAT-1 protein and mRNA levels were markedly reduced in DOC2b-enriched human islets. Intriguingly, DOC2b also prevents ER-stress-IKKβ and STAT-1 crosstalk in the rat INS1-832/13 β-cell line. Conclusion The mechanisms underpinning the protective effects of DOC2b involve attenuation of IKKβ-NF-κB p65 and STAT-1 signaling, and reduced CXCL10 expression. Graphical abstract
Collapse
|
5
|
Almutairi A, White TD, Stephenson DJ, Stephenson BD, Gai-Tusing Y, Goel P, Phillips DW, Welner RS, Lei X, Hammock BD, Chalfant CE, Ramanadham S. Selective Reduction of Ca2+-Independent Phospholipase A2β (iPLA2β)-Derived Lipid Signaling From Macrophages Mitigates Type 1 Diabetes Development. Diabetes 2024; 73:2022-2033. [PMID: 39283670 PMCID: PMC11579405 DOI: 10.2337/db23-0770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 09/07/2024] [Indexed: 11/22/2024]
Abstract
Type 1 diabetes (T1D) is a consequence of autoimmune destruction of β-cells, and macrophages (MΦs) have a central role in initiating processes that lead to β-cell demise. We reported that Ca2+-independent phospholipase A2β (iPLA2β)-derived lipid (iDL) signaling contributes to β-cell death. Because MΦs express iPLA2β, we assessed its role in T1D development. We find that selective reduction of myeloid-iPLA2β in spontaneously diabetes-prone NOD mice 1) decreases proinflammatory eicosanoid production by MΦs, 2) favors the anti-inflammatory (M2-like) MΦ phenotype, and 3) diminishes activated CD4+ and CD8+ T-cells phenotype in the pancreatic infiltrate, prior to T1D onset. These outcomes are associated with a significant reduction in T1D. Further, inhibition of select proinflammatory lipid signaling pathways reduces M1-like MΦ polarization and adoptive transfer of M2-like MΦs reduces NOD T1D incidence, suggesting a mechanism by which iDLs impact T1D development. These findings identify MΦ-iPLA2β as a critical contributor to T1D development and potential target to counter T1D onset. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Abdulaziz Almutairi
- Department of Cell, Developmental, and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
- Department of Basic Science, College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Tayleur D. White
- Department of Cell, Developmental, and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Daniel J. Stephenson
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA
| | - Benjamin D. Stephenson
- Program in Cancer Biology, UVA Comprehensive Cancer Center, University of Virginia School of Medicine, Charlottesville, VA
| | - Ying Gai-Tusing
- Department of Cell, Developmental, and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Paran Goel
- Department of Medicine, Hematology & Oncology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Daniel W. Phillips
- Department of Medicine, Hematology & Oncology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Robert S. Welner
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
- Department of Medicine, Hematology & Oncology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Xiaoyong Lei
- Department of Cell, Developmental, and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Bruce D. Hammock
- Department of Entomology and Nematology, University of California, Davis, Davis, CA
| | - Charles E. Chalfant
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA
- Program in Cancer Biology, UVA Comprehensive Cancer Center, University of Virginia School of Medicine, Charlottesville, VA
- Research Service, Richmond Veterans Administration Medical Center, Richmond, VA
| | - Sasanka Ramanadham
- Department of Cell, Developmental, and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
6
|
Auddino S, Aiello E, Grieco GE, Dotta F, Sebastiani G. A three-layer perspective on miRNA regulation in β cell inflammation. Trends Endocrinol Metab 2024:S1043-2760(24)00257-1. [PMID: 39532586 DOI: 10.1016/j.tem.2024.10.002] [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: 07/17/2024] [Revised: 09/10/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024]
Abstract
MicroRNAs (miRNAs) are noncoding RNA molecules that regulate gene expression post-transcriptionally and influence numerous biological processes. Aberrant miRNA expression is linked to diseases such as diabetes mellitus; indeed, miRNAs regulate pancreatic islet inflammation in both type 1 (T1D) and type 2 diabetes (T2D). Traditionally, miRNA research has focused on canonical sequences and offers a two-layer view - from expression to function. However, advances in RNA sequencing have revealed miRNA variants, called isomiRs, that arise from alternative processing or modifications of canonical sequences. This introduces a three-layer view - from expression, through sequence modifications, to function. We discuss the potential link between cellular stresses and isomiR biogenesis, and how this association could improve our knowledge of islet inflammation and dysfunction.
Collapse
Affiliation(s)
- Stefano Auddino
- Department of Medicine, Surgery, and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario Onlus, Toscana Life Sciences, Siena, Italy
| | - Elena Aiello
- Department of Medicine, Surgery, and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario Onlus, Toscana Life Sciences, Siena, Italy
| | - Giuseppina Emanuela Grieco
- Department of Medicine, Surgery, and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario Onlus, Toscana Life Sciences, Siena, Italy
| | - Francesco Dotta
- Department of Medicine, Surgery, and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario Onlus, Toscana Life Sciences, Siena, Italy; Tuscany Centre for Precision Medicine (CReMeP), Siena, Italy.
| | - Guido Sebastiani
- Department of Medicine, Surgery, and Neurosciences, University of Siena, Siena, Italy; Fondazione Umberto Di Mario Onlus, Toscana Life Sciences, Siena, Italy.
| |
Collapse
|
7
|
Holst-Hansen T, Nielsen PY, Jensen MH, Mandrup-Poulsen T, Trusina A. Tipping-point transition from transient to persistent inflammation in pancreatic islets. NPJ Syst Biol Appl 2024; 10:102. [PMID: 39266581 PMCID: PMC11393080 DOI: 10.1038/s41540-024-00427-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/19/2024] [Indexed: 09/14/2024] Open
Abstract
Type 2 diabetes (T2D) is associated with a systemic increase in the pro-inflammatory cytokine IL-1β. While transient exposure to low IL-1β concentrations improves insulin secretion and β-cell proliferation in pancreatic islets, prolonged exposure leads to impaired insulin secretion and collective β-cell death. IL-1 is secreted locally by islet-resident macrophages and β-cells; however, it is unknown if and how the two opposing modes may emerge at single islet level. We investigated the duality of IL-1β with a quantitative in silico model of the IL-1 regulatory network in pancreatic islets. We find that the network can produce either transient or persistent IL-1 responses when induced by pro-inflammatory and metabolic cues. This suggests that the duality of IL-1 may be regulated at the single islet level. We use two core feedbacks in the IL-1 regulation to explain both modes: First, a fast positive feedback in which IL-1 induces its own production through the IL-1R/IKK/NF-κB pathway. Second, a slow negative feedback where NF-κB upregulates inhibitors acting at different levels along the IL-1R/IKK/NF-κB pathway-IL-1 receptor antagonist and A20, among others. A transient response ensues when the two feedbacks are balanced. When the positive feedback dominates over the negative, islets transit into the persistent inflammation mode. Consistent with several observations, where the size of islets was implicated in its inflammatory state, we find that large islets and islets with high density of IL-1β amplifying cells are more prone to transit into persistent IL-1β mode. Our results are likely not limited to IL-1β but are general for the combined effect of multiple pro-inflammatory cytokines and chemokines. Generalizing complex regulations in terms of two feedback mechanisms of opposing nature and acting on different time scales provides a number of testable predictions. Taking islet architecture and cellular heterogeneity into consideration, further dynamic monitoring and experimental validation in actual islet samples will be crucial to verify the model predictions and enhance its utility in clinical applications.
Collapse
Affiliation(s)
| | - Pernille Yde Nielsen
- Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Lyngby, Denmark
| | - Mogens H Jensen
- Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark.
| | - Thomas Mandrup-Poulsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Ala Trusina
- Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
8
|
Zhang Y, Cong R, Lv T, Liu K, Chang X, Li Y, Han X, Zhu Y. Islet-resident macrophage-derived miR-155 promotes β cell decompensation via targeting PDX1. iScience 2024; 27:109540. [PMID: 38577099 PMCID: PMC10993184 DOI: 10.1016/j.isci.2024.109540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 02/18/2024] [Accepted: 03/18/2024] [Indexed: 04/06/2024] Open
Abstract
Chronic inflammation is critical for the initiation and progression of type 2 diabetes mellitus via causing both insulin resistance and pancreatic β cell dysfunction. miR-155, highly expressed in macrophages, is a master regulator of chronic inflammation. Here we show that blocking a macrophage-derived exosomal miR-155 (MDE-miR-155) mitigates the insulin resistances and glucose intolerances in high-fat-diet (HFD) feeding and type-2 diabetic db/db mice. Lentivirus-based miR-155 sponge decreases the level of miR-155 in the pancreas and improves glucose-stimulated insulin secretion (GSIS) ability of β cells, thus leading to improvements of insulin sensitivities in the liver and adipose tissues. Mechanistically, miR-155 increases its expression in HFD and db/db islets and is released as exosomes by islet-resident macrophages under metabolic stressed conditions. MDE-miR-155 enters β cells and causes defects in GSIS function and insulin biosynthesis via the miR-155-PDX1 axis. Our findings offer a treatment strategy for inflammation-associated diabetes via targeting miR-155.
Collapse
Affiliation(s)
- Yan Zhang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing medical University, Nanjing, Jiangsu 211166, China
| | - Rong Cong
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing medical University, Nanjing, Jiangsu 211166, China
| | - Tingting Lv
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing medical University, Nanjing, Jiangsu 211166, China
| | - Kerong Liu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing medical University, Nanjing, Jiangsu 211166, China
| | - Xiaoai Chang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing medical University, Nanjing, Jiangsu 211166, China
| | - Yating Li
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing medical University, Nanjing, Jiangsu 211166, China
| | - Xiao Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing medical University, Nanjing, Jiangsu 211166, China
| | - Yunxia Zhu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
9
|
Youssef N, Noureldein MH, Riachi ME, Haddad A, Eid AA. Macrophage polarization and signaling in diabetic kidney disease: a catalyst for disease progression. Am J Physiol Renal Physiol 2024; 326:F301-F312. [PMID: 38153850 DOI: 10.1152/ajprenal.00266.2023] [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: 08/30/2023] [Revised: 11/29/2023] [Accepted: 12/16/2023] [Indexed: 12/30/2023] Open
Abstract
Diabetic kidney disease (DKD) is a serious complication of diabetes affecting millions of people worldwide. Macrophages, a critical immune cell type, are central players in the development and progression of DKD. In this comprehensive review, we delve into the intricate role of macrophages in DKD, examining how they can become polarized into proinflammatory M1 or anti-inflammatory M2 phenotypes. We explore the signaling pathways involved in macrophage recruitment and polarization in the kidneys, including the key cytokines and transcription factors that promote M1 and M2 polarization. In addition, we discuss the latest clinical studies investigating macrophages in DKD and explore the potential of hypoglycemic drugs for modulating macrophage polarization. By gaining a deeper understanding of the mechanisms that regulate macrophage polarization in DKD, we may identify novel therapeutic targets for this debilitating complication of diabetes. This review provides valuable insights into the complex interplay between macrophages and DKD, shedding light on the latest developments in this important area of research. This review aims to enhance understanding of the role that macrophages play in the pathogenesis of DKD.
Collapse
Affiliation(s)
- Natalie Youssef
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- American University of Beirut Diabetes, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mohamed H Noureldein
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- American University of Beirut Diabetes, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mansour E Riachi
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- American University of Beirut Diabetes, American University of Beirut Medical Center, Beirut, Lebanon
| | - Antony Haddad
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- American University of Beirut Diabetes, American University of Beirut Medical Center, Beirut, Lebanon
| | - Assaad A Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- American University of Beirut Diabetes, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
10
|
Sionov RV, Ahdut-HaCohen R. A Supportive Role of Mesenchymal Stem Cells on Insulin-Producing Langerhans Islets with a Specific Emphasis on The Secretome. Biomedicines 2023; 11:2558. [PMID: 37761001 PMCID: PMC10527322 DOI: 10.3390/biomedicines11092558] [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: 08/15/2023] [Revised: 09/06/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Type 1 Diabetes (T1D) is a chronic autoimmune disease characterized by a gradual destruction of insulin-producing β-cells in the endocrine pancreas due to innate and specific immune responses, leading to impaired glucose homeostasis. T1D patients usually require regular insulin injections after meals to maintain normal serum glucose levels. In severe cases, pancreas or Langerhans islet transplantation can assist in reaching a sufficient β-mass to normalize glucose homeostasis. The latter procedure is limited because of low donor availability, high islet loss, and immune rejection. There is still a need to develop new technologies to improve islet survival and implantation and to keep the islets functional. Mesenchymal stem cells (MSCs) are multipotent non-hematopoietic progenitor cells with high plasticity that can support human pancreatic islet function both in vitro and in vivo and islet co-transplantation with MSCs is more effective than islet transplantation alone in attenuating diabetes progression. The beneficial effect of MSCs on islet function is due to a combined effect on angiogenesis, suppression of immune responses, and secretion of growth factors essential for islet survival and function. In this review, various aspects of MSCs related to islet function and diabetes are described.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- The Institute of Biomedical and Oral Research (IBOR), Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Ronit Ahdut-HaCohen
- Department of Medical Neurobiology, Institute of Medical Research, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel;
- Department of Science, The David Yellin Academic College of Education, Jerusalem 9103501, Israel
| |
Collapse
|
11
|
Gonuguntla S, Humphrey RK, Gorantla A, Hao E, Jhala US. Stress-induced pseudokinase TRB3 augments IL1β signaling by interacting with Flightless homolog 1. J Biol Chem 2023; 299:104803. [PMID: 37172723 PMCID: PMC10432976 DOI: 10.1016/j.jbc.2023.104803] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 04/20/2023] [Accepted: 04/22/2023] [Indexed: 05/15/2023] Open
Abstract
Interleukin-1β is one of the most potent inducers of beta cell inflammation in the lead-up to type 1 diabetes. We have previously reported that IL1β-stimulated pancreatic islets from mice with genetic ablation of stress-induced pseudokinase TRB3(TRB3KO) show attenuated activation kinetics for the MAP3K MLK3 and JNK stress kinases. However, JNK signaling constitutes only a portion of the cytokine-induced inflammatory response. Here we report that TRB3KO islets also show a decrease in amplitude and duration of IL1β-induced phosphorylation of TAK1 and IKK, kinases that drive the potent NF-κB proinflammatory signaling pathway. We observed that TRB3KO islets display decreased cytokine-induced beta cell death, preceded by a decrease in select downstream NF-κB targets, including iNOS/NOS2 (inducible nitric oxide synthase), a mediator of beta cell dysfunction and death. Thus, loss of TRB3 attenuates both pathways required for a cytokine-inducible, proapoptotic response in beta cells. In order to better understand the molecular basis of TRB3-enhanced, post-receptor IL1β signaling, we interrogated the TRB3 interactome using coimmunoprecipitation followed by mass spectrometry to identify immunomodulatory protein Flightless homolog 1 (Fli1) as a novel, TRB3-interacting protein. We show that TRB3 binds and disrupts Fli1-dependent sequestration of MyD88, thereby increasing availability of this most proximal adaptor required for IL1β receptor-dependent signaling. Fli1 sequesters MyD88 in a multiprotein complex resulting in a brake on the assembly of downstream signaling complexes. By interacting with Fli1, we propose that TRB3 lifts the brake on IL1β signaling to augment the proinflammatory response in beta cells.
Collapse
Affiliation(s)
- Sumati Gonuguntla
- Pediatric Diabetes Research Center, University of California San Diego, La Jolla, California, USA
| | - Rohan K Humphrey
- Pediatric Diabetes Research Center, University of California San Diego, La Jolla, California, USA
| | - Akshita Gorantla
- Pediatric Diabetes Research Center, University of California San Diego, La Jolla, California, USA
| | - Ergeng Hao
- Pediatric Diabetes Research Center, University of California San Diego, La Jolla, California, USA
| | - Ulupi S Jhala
- Pediatric Diabetes Research Center, University of California San Diego, La Jolla, California, USA.
| |
Collapse
|
12
|
Roca-Rivada A, Marín-Cañas S, Colli ML, Vinci C, Sawatani T, Marselli L, Cnop M, Marchetti P, Eizirik DL. Inhibition of the type 1 diabetes candidate gene PTPN2 aggravates TNF-α-induced human beta cell dysfunction and death. Diabetologia 2023; 66:1544-1556. [PMID: 36988639 DOI: 10.1007/s00125-023-05908-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 02/28/2023] [Indexed: 03/30/2023]
Abstract
AIMS/HYPOTHESIS TNF-α plays a role in pancreatic beta cell loss in type 1 diabetes mellitus. In clinical interventions, TNF-α inhibition preserves C-peptide levels in early type 1 diabetes. In this study we evaluated the crosstalk of TNF-α, as compared with type I IFNs, with the type 1 diabetes candidate gene PTPN2 (encoding protein tyrosine phosphatase non-receptor type 2 [PTPN2]) in human beta cells. METHODS EndoC-βH1 cells, dispersed human pancreatic islets or induced pluripotent stem cell (iPSC)-derived islet-like cells were transfected with siRNAs targeting various genes (siCTRL, siPTPN2, siJNK1, siJNK3 or siBIM). Cells were treated for 48 h with IFN-α (2000 U/ml) or TNF-α (1000 U/ml). Cell death was evaluated using Hoechst 33342 and propidium iodide staining. mRNA levels were assessed by quantitative reverse transcription PCR (qRT-PCR) and protein expression by immunoblot. RESULTS PTPN2 silencing sensitised beta cells to cytotoxicity induced by IFN-α and/or TNF-α by 20-50%, depending on the human cell model utilised; there was no potentiation between the cytokines. We silenced c-Jun N-terminal kinase (JNK)1 or Bcl-2-like protein 2 (BIM), and this abolished the proapoptotic effects of IFN-α, TNF-α or the combination of both after PTPN2 inhibition. We further observed that PTPN2 silencing increased TNF-α-induced JNK1 and BIM phosphorylation and that JNK3 is necessary for beta cell resistance to IFN-α cytotoxicity. CONCLUSIONS/INTERPRETATION We show that the type 1 diabetes candidate gene PTPN2 is a key regulator of the deleterious effects of TNF-α in human beta cells. It is conceivable that people with type 1 diabetes carrying risk-associated PTPN2 polymorphisms may particularly benefit from therapies inhibiting TNF-α.
Collapse
Affiliation(s)
- Arturo Roca-Rivada
- ULB Center for Diabetes Research, Medical Faculty, Université Libre De Bruxelles, Brussels, Belgium.
| | - Sandra Marín-Cañas
- ULB Center for Diabetes Research, Medical Faculty, Université Libre De Bruxelles, Brussels, Belgium
| | - Maikel L Colli
- ULB Center for Diabetes Research, Medical Faculty, Université Libre De Bruxelles, Brussels, Belgium
| | - Chiara Vinci
- ULB Center for Diabetes Research, Medical Faculty, Université Libre De Bruxelles, Brussels, Belgium
| | - Toshiaki Sawatani
- ULB Center for Diabetes Research, Medical Faculty, Université Libre De Bruxelles, Brussels, Belgium
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Miriam Cnop
- ULB Center for Diabetes Research, Medical Faculty, Université Libre De Bruxelles, Brussels, Belgium
- Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre De Bruxelles, Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
13
|
Oliveira VR, Paula CC, Taniguchi S, Ortis F. Pre-treatment with IL-6 potentiates β-cell death induced by pro-inflammatory cytokines. BMC Mol Cell Biol 2023; 24:11. [PMID: 36977992 PMCID: PMC10045109 DOI: 10.1186/s12860-023-00476-3] [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: 07/07/2022] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND Type I Diabetes mellitus (T1D) is characterized by a specific destruction of β-cells by the immune system. During this process pro-inflammatory cytokines are released in the pancreatic islets and contribute for β-cells demise. Cytokine-induced iNOS activation, via NF-κB, is implicated in induction of β-cells death, which includes ER stress activation. Physical exercise has been used as an adjunct for better glycemic control in patients with T1D, since it is able to increase glucose uptake independent of insulin. Recently, it was observed that the release of IL-6 by skeletal muscle, during physical exercise, could prevent β-cells death induced by pro-inflammatory cytokines. However, the molecular mechanisms involved in this beneficial effect on β-cells are not yet completely elucidated. Our aim was to evaluate the effect of IL-6 on β-cells exposed to pro-inflammatory cytokines. RESULTS Pre-treatment with IL-6 sensitized INS-1E cells to cytokine-induced cell death, increasing cytokine-induced iNOS and Caspase-3 expression. Under these conditions, however, there was a decrease in cytokines-induced p-eIF2-α but not p-IRE1expression, proteins related to ER stress. To address if this prevention of adequate UPR response is involved in the increase in β-cells death markers induced by IL-6 pre-treatment, we used a chemical chaperone (TUDCA), which improves ER folding capacity. Use of TUDCA increased cytokines-induced Caspase-3 expression and Bax/Bcl-2 ratio in the presence of IL-6 pre-treatment. However, there is no modulation of p-eIF2-α expression by TUDCA in this condition, with increase of CHOP expression. CONCLUSION Treatment with IL-6 alone is not beneficial for β-cells, leading to increased cell death markers and impaired UPR activation. In addition, TUDCA has not been able to restore ER homeostasis or improve β-cells viability under this condition, suggesting that other mechanisms may be involved.
Collapse
Affiliation(s)
- V R Oliveira
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - C C Paula
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - S Taniguchi
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - F Ortis
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
14
|
Malekpour M, Salarikia SR, Kashkooli M, Asadi-Pooya AA. The genetic link between systemic autoimmune disorders and temporal lobe epilepsy: A bioinformatics study. Epilepsia Open 2023. [PMID: 36929812 DOI: 10.1002/epi4.12727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/11/2023] [Indexed: 03/18/2023] Open
Abstract
OBJECTIVE We aimed to explore the underlying pathomechanisms of the comorbidity between three common systemic autoimmune disorders (SADs) [i.e., insulin-dependent diabetes mellitus (IDDM), systemic lupus erythematosus (SLE), and rheumatoid arthritis (RA)] and temporal lobe epilepsy (TLE), using bioinformatics tools. We hypothesized that there are shared genetic variations among these four conditions. METHODS Different databases (DisGeNET, Harmonizome, and Enrichr) were searched to find TLE-associated genes with variants; their single nucleotide polymorphisms (SNPs) were gathered from the literature. We also did a separate literature search using PubMed with the following keywords for original articles: "TLE" or "Temporal lobe epilepsy" AND "genetic variation," "single nucleotide polymorphism," "SNP," or "genetic polymorphism." In the next step, the SNPs associated with TLE were searched in the LitVar database to find the shared gene variations with RA, SLE, and IDDM. RESULTS Ninety unique SNPs were identified to be associated with TLE. LitVar search identified two SNPs that were shared between TLE and all three SADs (i.e., IDDM, SLE, and RA). The first SNP was rs16944 on the Interleukin-1β (IL-1β) gene. The second genetic variation was ε4 variation of apolipoprotein E (APOE) gene. SIGNIFICANCE The shared genetic variations (i.e., rs16944 on the IL-1β gene and ε4 variation of the APOE gene) may explain the underlying pathomechanisms of the comorbidity between three common SADs (i.e., IDDM, SLE, and RA) and TLE. Exploring such shared genetic variations may help find targeted therapies for patients with TLE, especially those with drug-resistant seizures who also have comorbid SADs.
Collapse
Affiliation(s)
- Mahdi Malekpour
- Epilepsy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mohammad Kashkooli
- Epilepsy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali A Asadi-Pooya
- Epilepsy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Neurology, Jefferson Comprehensive Epilepsy Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
15
|
Zheng W, Liu A, Xia N, Chen N, Meurens F, Zhu J. How the Innate Immune DNA Sensing cGAS-STING Pathway Is Involved in Apoptosis. Int J Mol Sci 2023; 24:3029. [PMID: 36769349 PMCID: PMC9917431 DOI: 10.3390/ijms24033029] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
The cGAS-STING signaling axis can be activated by cytosolic DNA, including both non-self DNA and self DNA. This axis is used by the innate immune system to monitor invading pathogens and/or damage. Increasing evidence has suggested that the cGAS-STING pathway not only facilitates inflammatory responses and the production of type I interferons (IFN), but also activates other cellular processes, such as apoptosis. Recently, many studies have focused on analyzing the mechanisms of apoptosis induced by the cGAS-STING pathway and their consequences. This review gives a detailed account of the interplay between the cGAS-STING pathway and apoptosis. The cGAS-STING pathway can induce apoptosis through ER stress, NLRP3, NF-κB, IRF3, and IFN signals. Conversely, apoptosis can feed back to regulate the cGAS-STING pathway, suppressing it via the activation of caspases or promoting it via mitochondrial DNA (mtDNA) release. Apoptosis mediated by the cGAS-STING pathway plays crucial roles in balancing innate immune responses, resisting infections, and limiting tumor growth.
Collapse
Affiliation(s)
- Wanglong Zheng
- College Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou 225009, China
- Comparative Medicine Research Institute, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Anjing Liu
- College Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou 225009, China
- Comparative Medicine Research Institute, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Nengwen Xia
- College Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou 225009, China
- Comparative Medicine Research Institute, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Nanhua Chen
- College Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou 225009, China
- Comparative Medicine Research Institute, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - François Meurens
- BIOEPAR, INRAE, Oniris, 44307 Nantes, France
- Department of Veterinary Microbiology and Immunology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
| | - Jianzhong Zhu
- College Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou 225009, China
- Comparative Medicine Research Institute, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| |
Collapse
|
16
|
Miao Z, Dong M, Wang Z, Ma J, Lin Y, Wu Y. Linalool inhibits the progression of osteoarthritis via the Nrf2/HO-1 signal pathway both in vitro and in vivo. Int Immunopharmacol 2022; 113:109338. [DOI: 10.1016/j.intimp.2022.109338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/28/2022] [Accepted: 10/09/2022] [Indexed: 11/05/2022]
|
17
|
Contreras CJ, Mukherjee N, Branco RCS, Lin L, Hogan MF, Cai EP, Oberst AA, Kahn SE, Templin AT. RIPK1 and RIPK3 regulate TNFα-induced β-cell death in concert with caspase activity. Mol Metab 2022; 65:101582. [PMID: 36030035 PMCID: PMC9464965 DOI: 10.1016/j.molmet.2022.101582] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 12/02/2022] Open
Abstract
OBJECTIVE Type 1 diabetes (T1D) is characterized by autoimmune-associated β-cell loss, insulin insufficiency, and hyperglycemia. Although TNFα signaling is associated with β-cell loss and hyperglycemia in non-obese diabetic mice and human T1D, the molecular mechanisms of β-cell TNF receptor signaling have not been fully characterized. Based on work in other cell types, we hypothesized that receptor interacting protein kinase 1 (RIPK1) and receptor interacting protein kinase 3 (RIPK3) regulate TNFα-induced β-cell death in concert with caspase activity. METHODS We evaluated TNFα-induced cell death, caspase activity, and TNF receptor pathway molecule expression in immortalized NIT-1 and INS-1 β-cell lines and primary mouse islet cells in vitro. Our studies utilized genetic and small molecule approaches to alter RIPK1 and RIPK3 expression and caspase activity to interrogate mechanisms of TNFα-induced β-cell death. We used the β-cell toxin streptozotocin (STZ) to determine the susceptibility of Ripk3+/+ and Ripk3-/- mice to hyperglycemia in vivo. RESULTS Expression of TNF receptor signaling molecules including RIPK1 and RIPK3 was identified in NIT-1 and INS-1 β cells and isolated mouse islets at the mRNA and protein levels. TNFα treatment increased NIT-1 and INS-1 cell death and caspase activity after 24-48 h, and BV6, a small molecule inhibitor of inhibitor of apoptosis proteins (IAPs) amplified this TNFα-induced cell death. RIPK1 deficient NIT-1 cells were protected from TNFα- and BV6-induced cell death and caspase activation. Interestingly, small molecule inhibition of caspases with zVAD-fmk (zVAD) did not prevent TNFα-induced cell death in either NIT-1 or INS-1 cells. This caspase-independent cell death was increased by BV6 treatment and decreased in RIPK1 deficient NIT-1 cells. RIPK3 deficient NIT-1 cells and RIPK3 kinase inhibitor treated INS-1 cells were protected from TNFα+zVAD-induced cell death, whereas RIPK3 overexpression increased INS-1 cell death and promoted RIPK3 and MLKL interaction under TNFα+zVAD treatment. In mouse islet cells, BV6 or zVAD treatment promoted TNFα-induced cell death, and TNFα+zVAD-induced cell death was blocked by RIPK3 inhibition and in Ripk3-/- islet cells in vitro. Ripk3-/- mice were also protected from STZ-induced hyperglycemia and glucose intolerance in vivo. CONCLUSIONS RIPK1 and RIPK3 regulate TNFα-induced β-cell death in concert with caspase activity in immortalized and primary islet β cells. TNF receptor signaling molecules such as RIPK1 and RIPK3 may represent novel therapeutic targets to promote β-cell survival and glucose homeostasis in T1D.
Collapse
Affiliation(s)
- Christopher J Contreras
- Division of Endocrinology, Department of Medicine, Roudebush VA Medical Center and Indiana University School of Medicine, Indianapolis, IN, USA
| | - Noyonika Mukherjee
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Renato C S Branco
- Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Li Lin
- Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Meghan F Hogan
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, WA, USA
| | - Erica P Cai
- Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Andrew A Oberst
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Steven E Kahn
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, WA, USA
| | - Andrew T Templin
- Division of Endocrinology, Department of Medicine, Roudebush VA Medical Center and Indiana University School of Medicine, Indianapolis, IN, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA; Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, Indianapolis, IN, USA; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
18
|
Shao S, Zhang Y, Li G, Yu Z, Cao Y, Zheng L, Zhang K, Han X, Shi Z, Cui H, Song X, Hong W, Han T. The dynamics of cell death patterns and regeneration during acute liver injury in mice. FEBS Open Bio 2022; 12:1061-1074. [PMID: 35184410 PMCID: PMC9063440 DOI: 10.1002/2211-5463.13383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/12/2021] [Accepted: 01/03/2022] [Indexed: 11/08/2022] Open
Abstract
Acute liver injury is a serious clinical syndrome with multiple causes and unclear pathological process. Here, CCl4‐ and D‐galactosamine/lipopolysaccharide (D‐gal/LPS)‐induced acute liver injury was established to explore the cell death patterns and determine whether or not liver regeneration occurred. In CCl4‐induced hepatic injury, three phases, including the early, progressive, and recovery phase, were considered based on alterations of serum transaminases and liver morphology. Moreover, in this model, cytokines exhibited double‐peak fluctuations; apoptosis and pyroptosis persisted throughout all phases; autophagy occurred in the early and the progressive phases; and sufficient and timely hepatocyte regeneration was observed only during the recovery phase. All of these phenomena contribute to mild liver injury and subsequent regeneration. Strikingly, only the early and progressive phases were observed in the D‐gal/LPS model. Slight pyroptosis occurred in the early phase but diminished in the progressive phase, while apoptosis, reduced autophagy, and slight but subsequently diminished regeneration occurred only during the progressive phase, accompanied by a strong cytokine storm, resulting in severe liver injury with high mortality. Taken together, our work reveals variable modes and dynamics of cell death and regeneration, which lead to different consequences for mild and severe acute liver injury, providing a helpful reference for clinical therapy and prognosis.
Collapse
Affiliation(s)
- Shuai Shao
- The School of Medicine NanKai University Tianjin China
| | - Yu Zhang
- Department of Hepatology and Gastroenterology The Third Central Clinical College of Tianjin Medical University Department of Histology and Embryology School of Basic Medical Sciences Tianjin Medical University China
| | - Guantong Li
- Department of Hepatology and Gastroenterology The Third Central Clinical College of Tianjin Medical University Department of Histology and Embryology School of Basic Medical Sciences Tianjin Medical University China
| | - Zhenjun Yu
- Department of Hepatology and Gastroenterology The Third Central Clinical College of Tianjin Medical University Department of Histology and Embryology School of Basic Medical Sciences Tianjin Medical University China
| | - Yingying Cao
- Department of Hepatology and Gastroenterology The Third Central Clinical College of Tianjin Medical University Department of Histology and Embryology School of Basic Medical Sciences Tianjin Medical University China
| | - Lina Zheng
- Department of Histology and Embryology School of Basic Medical Sciences Tianjin Medical University Tianjin China
| | - Kun Zhang
- Department of Histology and Embryology School of Basic Medical Sciences Tianjin Medical University Tianjin China
| | - Xiaohui Han
- Department of Histology and Embryology School of Basic Medical Sciences Tianjin Medical University Tianjin China
| | - Zhemin Shi
- Department of Histology and Embryology School of Basic Medical Sciences Tianjin Medical University Tianjin China
| | - Hongmei Cui
- Department of Histology and Embryology School of Basic Medical Sciences Tianjin Medical University Tianjin China
| | - Xiaomeng Song
- Department of Histology and Embryology School of Basic Medical Sciences Tianjin Medical University Tianjin China
| | - Wei Hong
- Department of Histology and Embryology School of Basic Medical Sciences Tianjin Medical University Tianjin China
| | - Tao Han
- The School of Medicine NanKai University Tianjin China
- Department of Hepatology and Gastroenterology The Third Central Clinical College of Tianjin Medical University Department of Histology and Embryology School of Basic Medical Sciences Tianjin Medical University China
- Department of Hepatology and Gastroenterology Tianjin Union Medical Center Nankai University Tianjin China
- Department of Hepatology and Gastroenterology Tianjin Third Central Hospital affiliated to Nankai University Tianjin China
| |
Collapse
|
19
|
Yang Y, Chen Z, Zhao X, Xie H, Du L, Gao H, Xie C. Mechanisms of Kaempferol in the treatment of diabetes: A comprehensive and latest review. Front Endocrinol (Lausanne) 2022; 13:990299. [PMID: 36157449 PMCID: PMC9490412 DOI: 10.3389/fendo.2022.990299] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/17/2022] [Indexed: 01/07/2023] Open
Abstract
Obesity-insulin resistance-β-cells apoptosis" is an important trilogy of the pathogenesis of type 2 diabetes. With the global pandemic of obesity and diabetes, continuous research and development of new drugs focuses on the prevention of the pathological progress of these diseases. According to a recent study, the natural product kaempferol has excellent antidiabetic effects. Therefore, this review comprehensively summarized the frontier studies and pharmacological mechanisms of kaempferol in the treatment of diabetes. The successful research and development of kaempferol may yield a significant leap in the treatment of diabetes and its complications.
Collapse
Affiliation(s)
- Yan Yang
- Hospital of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Zhengtao Chen
- Hospital of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoyan Zhao
- Hospital of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Hongyan Xie
- Hospital of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Lian Du
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong Gao
- Hospital of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Hong Gao, ; Chunguang Xie,
| | - Chunguang Xie
- Hospital of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Hong Gao, ; Chunguang Xie,
| |
Collapse
|
20
|
Murata T, Hashimoto K, Kohno S, Takahashi C, Yamaguchi M, Ito C, Masataka I, Kojima R, Hikita K, Kaneda N. Chemical inducer of regucalcin attenuates lipopolysaccharide-induced inflammatory responses in pancreatic MIN6 β-cells and RAW264.7 macrophages. FEBS Open Bio 2022; 12:175-191. [PMID: 34709731 PMCID: PMC8727933 DOI: 10.1002/2211-5463.13321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 07/28/2021] [Accepted: 10/27/2021] [Indexed: 11/27/2022] Open
Abstract
We previously isolated derrisfolin A, a novel rotenoid derivative, from the stems of Derris trifoliata Lour. (Leguminosae). Here, we report that derrisfolin A induces the expression of endogenous regucalcin (RGN) protein in both pancreatic MIN6 β-cells and RAW264.7 macrophages. Induction of RGN expression by derrisfolin A or retrovirus-mediated gene transfer in MIN6 cells and RAW264.7 macrophages significantly decreased lipopolysaccharide (LPS)-induced mRNA expression of Nos2, Il1b, and Tnf via nuclear factor-κB activation; reduced LPS-induced apoptosis in MIN6 cells, accompanied by decreased production of nitric oxide, interleukin-1β, and tumor necrosis factor-α; and attenuated generation of LPS-induced reactive oxygen species, malondialdehyde, and 3-nitrotyrosine in MIN6 cells. Additionally, in co-cultures of MIN6 cells with RAW264.7 macrophages in the presence of LPS, induction of RGN expression by derrisfolin A or retrovirus-mediated gene transfer in RAW264.7 macrophages attenuated apoptosis and oxidative/nitrosative stress in MIN6 cells. These results suggest that the induction of RGN expression in MIN6 cells was effective in suppressing LPS-induced inflammatory cytotoxicity and that in co-culture conditions, the induction of RGN expression in RAW264.7 macrophages blocked LPS-induced paracrine effects of RAW264.7 macrophages on inflammatory cytotoxicity in MIN6 cells. Our findings suggest that derrisfolin A, a chemical inducer of RGN, might be useful for developing a new drug against macrophage-associated β-cell inflammation in type 2 diabetes.
Collapse
Affiliation(s)
- Tomiyasu Murata
- Laboratory of Molecular BiologyFaculty of PharmacyMeijo UniversityNagoyaJapan
| | - Kazunori Hashimoto
- Laboratory of Molecular BiologyFaculty of PharmacyMeijo UniversityNagoyaJapan
| | - Susumu Kohno
- Division of Oncology and Molecular BiologyCancer Research InstituteKanazawa UniversityJapan
| | - Chiaki Takahashi
- Division of Oncology and Molecular BiologyCancer Research InstituteKanazawa UniversityJapan
| | - Masayoshi Yamaguchi
- Cancer Biology ProgramUniversity of Hawaii Cancer CenterUniversity of Hawaii at ManoaHonoluluHIUSA
| | - Chihiro Ito
- Laboratory of Natural Products ChemistryFaculty of PharmacyMeijo UniversityNagoyaJapan
| | - Itoigawa Masataka
- School of Sport and Health ScienceTokai Gakuen UniversityMiyoshiJapan
| | - Roji Kojima
- Laboratory of Analytical PharmacyFaculty of PharmacyMeijo UniversityNagoyaJapan
| | - Kiyomi Hikita
- Department of PharmacyFaculty of PharmacyGifu University of Medical ScienceKaniJapan
| | - Norio Kaneda
- Laboratory of Molecular BiologyFaculty of PharmacyMeijo UniversityNagoyaJapan
| |
Collapse
|
21
|
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]
|
22
|
Shazmeen, Haq I, Rajoka MSR, Asim Shabbir M, Umair M, llah I, Manzoor MF, Nemat A, Abid M, Khan MR, Aadil RM. Role of stilbenes against insulin resistance: A review. Food Sci Nutr 2021; 9:6389-6405. [PMID: 34760269 PMCID: PMC8565239 DOI: 10.1002/fsn3.2553] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/07/2021] [Accepted: 08/14/2021] [Indexed: 12/29/2022] Open
Abstract
Insulin resistance (IR) is a state characterized by the inability of tissues to utilize blood glucose particularly liver, muscle, and adipose tissues resulting in hyperglycemia and hyperinsulinemia. A close relationship exists between IR and the development of type 2 diabetes (T2D). Therefore, therapeutic approaches to treat IR also improve T2D simultaneously. Scientific evidence has highlighted the major role of inflammatory cytokines, reactive oxygen species (ROS), environmental & genetic factors, and auto-immune disorders in the pathophysiology of IR. Among therapeutic remedies, nutraceuticals like polyphenols are being used widely to ameliorate IR due to their safer nature compared to pharmaceutics. Stilbenes are considered important metabolically active polyphenols currently under the limelight of research to cope with IR. In this review, efforts are made to elucidate cellular and subcellular mechanisms influenced by stilbenes including modulating insulin signaling cascade, correcting glucose transport pathways, lowering postprandial glucose levels, and protecting β-cell damage and its effects on the hyperactive immune system and proinflammatory cytokines to attenuate IR. Furthermore, future directions to further the research in stilbenes as a strong candidate against IR are included so that concrete recommendation for their use in humans is made.
Collapse
Affiliation(s)
- Shazmeen
- National Institute of Food Science and TechnologyUniversity of AgricultureFaisalabadPakistan
| | - Iahtisham‐Ul Haq
- School of Food and NutritionFaculty of Allied Health SciencesMinhaj UniversityLahorePakistan
| | - Muhammad Shahid Riaz Rajoka
- Food and Feed Immunology GroupLaboratory of Animal Food FunctionGraduate School of Agricultural ScienceTohoku UniversitySendaiJapan
| | - Muhmmad Asim Shabbir
- National Institute of Food Science and TechnologyUniversity of AgricultureFaisalabadPakistan
| | - Muhammad Umair
- Department of Food Science and EngineeringCollege of Chemistry and EngineeringShenzhen UniversityShenzhenChina
| | - Inam‐u llah
- Department of Food Science and TechnologyThe University of HaripurKhyber‐PakhtunkhwaPakistan
| | - Muhammad Faisal Manzoor
- School of Food and Biological EngineeringJiangsu UniversityZhenjiangChina
- Riphah College of Rehabilitation and Allied Health SciencesRiphah International UniversityFaisalabadPakistan
| | - Arash Nemat
- Department of MicrobiologyKabul University of Medical SciencesKabulAfghanistan
| | - Muhammad Abid
- Institute of Food and Nutritional SciencesArid Agriculture UniversityRawalpindiPakistan
| | - Moazzam Rafiq Khan
- National Institute of Food Science and TechnologyUniversity of AgricultureFaisalabadPakistan
| | - Rana Muhammad Aadil
- National Institute of Food Science and TechnologyUniversity of AgricultureFaisalabadPakistan
| |
Collapse
|
23
|
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: 13] [Impact Index Per Article: 3.3] [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
|
24
|
Cosentino C, Regazzi R. Crosstalk between Macrophages and Pancreatic β-Cells in Islet Development, Homeostasis and Disease. Int J Mol Sci 2021; 22:ijms22041765. [PMID: 33578952 PMCID: PMC7916718 DOI: 10.3390/ijms22041765] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/04/2021] [Accepted: 02/06/2021] [Indexed: 12/29/2022] Open
Abstract
Macrophages are highly heterogeneous and plastic immune cells with peculiar characteristics dependent on their origin and microenvironment. Following pathogen infection or damage, circulating monocytes can be recruited in different tissues where they differentiate into macrophages. Stimuli present in the surrounding milieu induce the polarisation of macrophages towards a pro-inflammatory or anti-inflammatory profile, mediating inflammatory or homeostatic responses, respectively. However, macrophages can also derive from embryonic hematopoietic precursors and reside in specific tissues, actively participating in the development and the homeostasis in physiological conditions. Pancreatic islet resident macrophages are present from the prenatal stages onwards and show specific surface markers and functions. They localise in close proximity to β-cells, being exquisite sensors of their secretory ability and viability. Over the years, the crucial role of macrophages in β-cell differentiation and homeostasis has been highlighted. In addition, macrophages are emerging as central players in the initiation of autoimmune insulitis in type 1 diabetes and in the low-grade chronic inflammation characteristic of obesity and type 2 diabetes pathogenesis. The present work reviews the current knowledge in the field, with a particular focus on the mechanisms of communication between β-cells and macrophages that have been described so far.
Collapse
Affiliation(s)
- Cristina Cosentino
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, CH-1005 Lausanne, Switzerland;
| | - Romano Regazzi
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, CH-1005 Lausanne, Switzerland;
- Department of Biomedical Sciences, University of Lausanne, Rue du Bugnon 7, CH-1005 Lausanne, Switzerland
- Correspondence: ; Tel.: +41-21-692-52-80; Fax: +41-21-692-52-55
| |
Collapse
|
25
|
Pang H, Luo S, Huang G, Xia Y, Xie Z, Zhou Z. Advances in Knowledge of Candidate Genes Acting at the Beta-Cell Level in the Pathogenesis of T1DM. Front Endocrinol (Lausanne) 2020; 11:119. [PMID: 32226409 PMCID: PMC7080653 DOI: 10.3389/fendo.2020.00119] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 02/24/2020] [Indexed: 12/12/2022] Open
Abstract
T1DM (type 1 diabetes mellitus), which results from the irreversible elimination of beta-cells mediated by autoreactive T cells, is defined as an autoimmune disease. It is widely accepted that T1DM is caused by a combination of genetic and environmental factors, but the precise underlying molecular mechanisms are still unknown. To date, more than 50 genetic risk regions contributing to the pathogenesis of T1DM have been identified by GWAS (genome-wide association studies). Notably, more than 60% of the identified candidate genes are expressed in islets and beta-cells, which makes it plausible that these genes act at the beta-cell level and play a key role in the pathogenesis of T1DM. In this review, we focus on the current status of candidate genes that act at the beta-cell level by regulating the innate immune response and antiviral activity, affecting susceptibility to proapoptotic stimuli and influencing the pancreatic beta-cell phenotype.
Collapse
Affiliation(s)
- Haipeng Pang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| | - Shuoming Luo
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| | - Gan Huang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| | - Ying Xia
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| | - Zhiguo Xie
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
- *Correspondence: Zhiguo Xie
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
- Zhiguang Zhou
| |
Collapse
|
26
|
Song Z, Ma J, Lu Y, Zhou C, Zhao T, Ai X, Wei X, Lin J, Wang W, Yan W, Jiao P. The protective role of the MKP-5-JNK/P38 pathway in glucolipotoxicity-induced islet β-cell dysfunction and apoptosis. Exp Cell Res 2019; 382:111467. [DOI: 10.1016/j.yexcr.2019.06.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 06/10/2019] [Accepted: 06/11/2019] [Indexed: 12/12/2022]
|
27
|
Yang C, He L, Wang C, Huang Y, Wang A, Li X, Ao J. Dexmedetomidine alleviated lipopolysaccharide/D-galactosamine-induced acute liver injury in mice. Int Immunopharmacol 2019; 72:367-373. [DOI: 10.1016/j.intimp.2019.04.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 03/29/2019] [Accepted: 04/08/2019] [Indexed: 01/29/2023]
|
28
|
The Association between Depression and Type 1 Diabetes Mellitus: Inflammatory Cytokines as Ferrymen in between? Mediators Inflamm 2019; 2019:2987901. [PMID: 31049023 PMCID: PMC6458932 DOI: 10.1155/2019/2987901] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 02/14/2019] [Indexed: 12/16/2022] Open
Abstract
The depression incidence is much higher in patients with diabetes mellitus (DM), and the majority of these cases remain under-diagnosed. Type 1 diabetes mellitus (T1D) is now widely thought to be an organ-specific autoimmune disease. As a chronic autoimmune condition, T1D is characterized by T cell-mediated selective loss of insulin-producing β-cells. The age of onset of T1D is earlier than T2D, and T1D patients have an increased vulnerability to depression due to its diagnosis and treatment burden occurring in a period when the individuals are young. The literature has suggested that inflammatory cytokines play a wide role in both diseases. In this review, the mechanisms behind the initiation and propagation of the autoimmune response in T1D and depression are analyzed, and the contribution of cytokines to both conditions is discussed. This review outlines the immunological mechanism of T1D and depression, with a particular emphasis on the role of tumor necrosis factor-α (TNF-α), IL-1β, and interferon-γ (IFN-γ) cytokines and their signaling pathways. The purpose of this review is to highlight the possible pathways of the cytokines shared by these two diseases via deciphering their cytokine cascades. They may provide a basic groundwork for future study of the possible mechanism that links these two diseases and to develop new compounds that target the same pathway but can conquer two diseases.
Collapse
|
29
|
Tyka K, Jörns A, Turatsinze JV, Eizirik DL, Lenzen S, Gurgul-Convey E. MCPIP1 regulates the sensitivity of pancreatic beta-cells to cytokine toxicity. Cell Death Dis 2019; 10:29. [PMID: 30631045 PMCID: PMC6328635 DOI: 10.1038/s41419-018-1268-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 10/29/2018] [Accepted: 12/05/2018] [Indexed: 12/20/2022]
Abstract
The autoimmune-mediated beta-cell death in type 1 diabetes (T1DM) is associated with local inflammation (insulitis). We examined the role of MCPIP1 (monocyte chemotactic protein–induced protein 1), a novel cytokine-induced antiinflammatory protein, in this process. Basal MCPIP1 expression was lower in rat vs. human islets and beta-cells. Proinflammatory cytokines stimulated MCPIP1 expression in rat and human islets and in insulin-secreting cells. Moderate overexpression of MCPIP1 protected insulin-secreting INS1E cells against cytokine toxicity by a mechanism dependent on the presence of the PIN/DUB domain in MCPIP1. It also reduced cytokine-induced Chop and C/ebpβ expression and maintained MCL-1 expression. The shRNA-mediated suppression of MCPIP1 led to the potentiation of cytokine-mediated NFκB activation and cytokine toxicity in human EndoC-βH1 beta-cells. MCPIP1 expression was very high in infiltrated beta-cells before and after diabetes manifestation in the LEW.1AR1-iddm rat model of human T1DM. The extremely high expression of MCPIP1 in clonal beta-cells was associated with a failure of the regulatory feedback-loop mechanism, ER stress induction and high cytokine toxicity. In conclusion, our data indicate that the expression level of MCPIP1 affects the susceptibility of insulin-secreting cells to cytokines and regulates the mechanism of beta-cell death in T1DM.
Collapse
Affiliation(s)
- Karolina Tyka
- Institute of Clinical Biochemistry, Hannover Medical School, 30625, Hannover, Germany
| | - Anne Jörns
- Institute of Clinical Biochemistry, Hannover Medical School, 30625, Hannover, Germany
| | - Jean-Valery Turatsinze
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Sigurd Lenzen
- Institute of Clinical Biochemistry, Hannover Medical School, 30625, Hannover, Germany.,Institute of Experimental Diabetes Research, Hannover Medical School, Hannover, Germany
| | - Ewa Gurgul-Convey
- Institute of Clinical Biochemistry, Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
30
|
Oh E, Ahn M, Afelik S, Becker TC, Roep BO, Thurmond DC. Syntaxin 4 Expression in Pancreatic β-Cells Promotes Islet Function and Protects Functional β-Cell Mass. Diabetes 2018; 67:2626-2639. [PMID: 30305365 PMCID: PMC6245223 DOI: 10.2337/db18-0259] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 09/25/2018] [Indexed: 02/06/2023]
Abstract
Syntaxin 4 (Stx4) enrichment in human and mouse islet grafts improves the success of transplants in reversing streptozotocin (STZ)-induced diabetes in mice, although the underlying molecular mechanisms remain elusive. Toward a further understanding of this, human islets and inducible transgenic mice that selectively overexpress Stx4 in islet β-cells (βTG-Stx4) were challenged with proinflammatory stressors in vitro and in vivo. Remarkably, βTG-Stx4 mice resisted the loss of β-cell mass and the glucose intolerance that multiple low doses of STZ induce. Under standard conditions, glucose tolerance was enhanced and mice maintained normal fasting glycemia and insulinemia. Conversely, Stx4 heterozygous knockout mice succumbed rapidly to STZ-induced glucose intolerance compared with their wild-type littermates. Human islet β-cells overexpressing Stx4 exhibited enhanced insulin secretory capability; resilience against proinflammatory cytokine-induced apoptosis; and reduced expression of the CXCL9, CXCL10, and CXCL11 genes coordinate with decreased activation/nuclear localization of nuclear factor-κB. Finding ways to boost Stx4 expression presents a novel potential therapeutic avenue for promoting islet function and preserving β-cell mass.
Collapse
Affiliation(s)
- Eunjin Oh
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolism Research Institute of City of Hope, Duarte, CA
| | - Miwon Ahn
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolism Research Institute of City of Hope, Duarte, CA
| | - Solomon Afelik
- Department of Surgery/Division of Transplantation, University of Illinois at Chicago, Chicago, IL
| | - Thomas C Becker
- Department of Internal Medicine, Duke Molecular Physiology Institute, Duke University, Durham, NC
| | - Bart O Roep
- Department of Diabetes Immunology, Diabetes and Metabolism Research Institute of City of Hope, Duarte, CA
| | - Debbie C Thurmond
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolism Research Institute of City of Hope, Duarte, CA
| |
Collapse
|
31
|
Nardelli TR, Vanzela EC, Benedicto KC, Brozzi F, Fujita A, Cardozo AK, Eizirik DL, Boschero AC, Ortis F. Prolactin protects against cytokine-induced beta-cell death by NFκB and JNK inhibition. J Mol Endocrinol 2018; 61:25-36. [PMID: 29632026 DOI: 10.1530/jme-16-0257] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 04/09/2018] [Indexed: 12/25/2022]
Abstract
Type 1 diabetes is caused by an autoimmune assault that induces progressive beta-cell dysfunction and dead. Pro-inflammatory cytokines, such as interleukin 1 beta (IL1B), tumor necrosis factor (TNF) and interferon gamma (IFNG) contribute for beta-cell death, which involves the activation of the nuclear factor kappa B (NFκB) and c- Jun N-terminal kinase (JNK). Prolactin (PRL), a physiological mediator for beta-cell proliferation, was shown to protect beta cells against cytokines pro-apoptotic effects. We presently investigated the mechanisms involved in the protective effects of prolactin against cytokine-induced beta-cell death. The findings obtained indicate that STAT3 activation is involved in the anti-apoptotic role of PRL in rat beta cells. PRL prevents the activation of JNK via AKT and promotes a shift from expression of pro- to anti-apoptotic proteins downstream of the JNK cascade. Furthermore, PRL partially prevents the activation of NFκB and the transcription of its target genes IkBa, Fas, Mcp1, A20 and Cxcl10 and also decreases NO production. On the other hand, the pro-survival effects of PRL do not involve modulation of cytokine-induced endoplasmic reticulum stress. These results suggest that the beneficial effects of PRL in beta cells involve augmentation of anti-apoptotic mechanisms and, at the same time, reduction of pro-apoptotic effectors, rendering beta cells better prepared to deal with inflammatory insults. The better understanding of the pro-survival mechanisms modulated by PRL in beta cells can provide tools to prevent cell demise during an autoimmune attack or following islet transplantation.
Collapse
Affiliation(s)
- Tarlliza R Nardelli
- Department of Structural and Functional Biology, Laboratory of Endocrine Pancreas and Metabolism, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Emerielle C Vanzela
- Department of Structural and Functional Biology, Laboratory of Endocrine Pancreas and Metabolism, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Keli C Benedicto
- Department of Structural and Functional Biology, Laboratory of Endocrine Pancreas and Metabolism, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Flora Brozzi
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - André Fujita
- Department of Computer Science, Institute of Mathematics and Statistics, University of São Paulo (USP), São Paulo, Brazil
| | - Alessandra K Cardozo
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Décio L Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Antonio C Boschero
- Department of Structural and Functional Biology, Laboratory of Endocrine Pancreas and Metabolism, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Fernanda Ortis
- Department of Cell and Developmental Biology, Institute of Biomedical Science (ICB), University of São Paulo (USP), São Paulo, Brazil
| |
Collapse
|
32
|
Zhou H, Tang L, Yang Y, Lin L, Dai J, Ge P, Ai Q, Jiang R, Zhang L. Dopamine alleviated acute liver injury induced by lipopolysaccharide/d-galactosamine in mice. Int Immunopharmacol 2018; 61:249-255. [PMID: 29894864 DOI: 10.1016/j.intimp.2018.06.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 05/31/2018] [Accepted: 06/04/2018] [Indexed: 12/19/2022]
Abstract
Dopamine (DA), a crucial neurotransmitter, not only functions in the central nervous system but also plays important roles in the modulation of inflammation. Several studies suggest that DA might suppress the inflammatory response both in vitro and in vivo. In the present study, the potential effects of DA in a mouse model with lipopolysaccharide (LPS)/d-galactosamine (D-Gal)-induced acute liver injury were investigated. The results show that DA-treated LPS/D-Gal-exposed mice had reduced incidence of histologic lesions, lower plasma aminotransferases and improved the survival rates compared to LPS/D-Gal-exposed mice. Treatment with DA also suppressed LPS/D-Gal-induced production of TNF-α, phosphorylation of c-jun-N-terminal kinase (JNK), cleavage of caspase-3, up-regulation of hepatic caspase-3, caspase-8, and caspase-9 activities and reduced the count of TUNEL-positive hepatocytes. These data indicate that DA attenuated LPS/D-Gal-induced fulminant liver injury in mice, which implies that DA might have value for the prevention of inflammatory liver disease.
Collapse
Affiliation(s)
- Honghong Zhou
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Li Tang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Yongqiang Yang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Ling Lin
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Jie Dai
- Hospital of Chongqing University of Arts and Sciences, Chongqing, China
| | - Pu Ge
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Qing Ai
- Department of Physiology, Chongqing Medical University, Chongqing, China
| | - Rong Jiang
- Laboratory of Stem cell and Tissue Engineering, Chongqing Medical University, Chongqing, China
| | - Li Zhang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China; Laboratory of Stem cell and Tissue Engineering, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
33
|
Park JW, Lee HS, Lim Y, Paik JH, Kwon OK, Kim JH, Paryanto I, Yunianto P, Choi S, Oh SR, Ahn KS. Rhododendron album Blume extract inhibits TNF-α/IFN-γ-induced chemokine production via blockade of NF-κB and JAK/STAT activation in human epidermal keratinocytes. Int J Mol Med 2018. [PMID: 29532855 DOI: 10.3892/ijmm.2018.3556] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Rhododendron album Blume (RA) has traditionally been used as an herbal medicine and is considered to have anti‑inflammatory properties. It is a well‑known medicine for treatment of allergic or atopic diseases. In the present study, the biological effects of an RA methanol extract (RAME) on inflammation were investigated in tumor necrosis factor‑α (TNF‑α)/interferon‑γ (IFN‑γ)‑stimulated human keratinocytes. The present study aimed to investigate the potential mechanisms by which RAME inhibited TNF‑α/IFN‑γ‑induced expression of chemokines [thymus‑ and activation-regulated chemokine (TARC) and macrophage‑derived chemokine (MDC)] and cytokines [interleukin (IL)‑6 and IL‑8] through the nuclear factor‑κB (NF‑κB) pathway in human keratinocytes. The effects of RAME treatment on cell viability were investigated in TNF‑α/IFN‑γ‑stimulated HaCaT cells. The expression of TARC, MDC, IL‑6 and IL‑8 was assessed using reverse transcription‑quantitative polymerase chain reaction analysis or ELISA, and its effect on the inhibitory mitogen-activated protein kinase pathway was also studied using western blot analysis. TNF‑α/IFN‑γ induced the expression of IL‑6, IL‑8, TARC and MDC in a dose‑dependent manner through NF‑κB and Janus kinase/signal transducers and activators of transcription (JAK/STAT) activation. Notably, treatment with RAME significantly suppressed TNF-α/IFN-γ-induced expression of IL‑6, IL‑8, TARC, and MDC. In addition, RAME treatment inhibited the activation of NF‑κB and the JAK/STAT pathway in TNF‑α/IFN‑γ‑induced HaCaT cells. These results suggest that RAME decreases the production of chemokines and pro‑inflammatory cytokines by suppressing the NF‑κB and the JAK/STAT pathways. Consequently, RAME may potentially be used for treatment of atopic dermatitis.
Collapse
Affiliation(s)
- Ji-Won Park
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungbuk 28116, Republic of Korea
| | - Han-Sol Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungbuk 28116, Republic of Korea
| | - Yourim Lim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungbuk 28116, Republic of Korea
| | - Jin-Hyub Paik
- International Biological Material Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Ok-Kyoung Kwon
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungbuk 28116, Republic of Korea
| | - Jung-Hee Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungbuk 28116, Republic of Korea
| | - Imam Paryanto
- Center for Pharmaceutical and Medical Technology, Kawasan Puspiptek Serpong, LAPTIAB, Tangerang, Banten 15314, Indonesia
| | - Prasetyawan Yunianto
- Center for Pharmaceutical and Medical Technology, Kawasan Puspiptek Serpong, LAPTIAB, Tangerang, Banten 15314, Indonesia
| | - Sangho Choi
- International Biological Material Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungbuk 28116, Republic of Korea
| | - Kyung-Seop Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungbuk 28116, Republic of Korea
| |
Collapse
|
34
|
Kaminitz A, Ash S, Askenasy N. Neutralization Versus Reinforcement of Proinflammatory Cytokines to Arrest Autoimmunity in Type 1 Diabetes. Clin Rev Allergy Immunol 2018; 52:460-472. [PMID: 27677500 DOI: 10.1007/s12016-016-8587-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
As physiological pathways of intercellular communication produced by all cells, cytokines are involved in the pathogenesis of inflammatory insulitis as well as pivotal mediators of immune homeostasis. Proinflammatory cytokines including interleukins, interferons, transforming growth factor-β, tumor necrosis factor-α, and nitric oxide promote destructive insulitis in type 1 diabetes through amplification of the autoimmune reaction, direct toxicity to β-cells, and sensitization of islets to apoptosis. The concept that neutralization of cytokines may be of therapeutic benefit has been tested in few clinical studies, which fell short of inducing sustained remission or achieving disease arrest. Therapeutic failure is explained by the redundant activities of individual cytokines and their combinations, which are rather dispensable in the process of destructive insulitis because other cytolytic pathways efficiently compensate their deficiency. Proinflammatory cytokines are less redundant in regulation of the inflammatory reaction, displaying protective effects through restriction of effector cell activity, reinforcement of suppressor cell function, and participation in islet recovery from injury. Our analysis suggests that the role of cytokines in immune homeostasis overrides their contribution to β-cell death and may be used as potent immunomodulatory agents for therapeutic purposes rather than neutralized.
Collapse
Affiliation(s)
- Ayelet Kaminitz
- The Leah and Edward M. Frankel Laboratory of Experimental Bone Marrow Transplantation, 14 Kaplan Street, Petach Tikva, Israel, 49202
| | - Shifra Ash
- The Leah and Edward M. Frankel Laboratory of Experimental Bone Marrow Transplantation, 14 Kaplan Street, Petach Tikva, Israel, 49202
| | - Nadir Askenasy
- The Leah and Edward M. Frankel Laboratory of Experimental Bone Marrow Transplantation, 14 Kaplan Street, Petach Tikva, Israel, 49202.
| |
Collapse
|
35
|
Yamada S, Guo X. Peroxiredoxin 4 (PRDX4): Its critical in vivo
roles in animal models of metabolic syndrome ranging from atherosclerosis to nonalcoholic fatty liver disease. Pathol Int 2018; 68:91-101. [PMID: 29341349 DOI: 10.1111/pin.12634] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 12/13/2017] [Indexed: 01/07/2023]
Affiliation(s)
- Sohsuke Yamada
- Department of Pathology and Laboratory Medicine; Kanazawa Medical University; Ishikawa Japan
| | - Xin Guo
- Department of Pathology and Laboratory Medicine; Kanazawa Medical University; Ishikawa Japan
- Laboratory of Pathology; Hebei Cancer Institute; The Fourth Hospital of Hebei Medical University; Hebei China
| |
Collapse
|
36
|
Yoshimatsu G, Kunnathodi F, Saravanan PB, Shahbazov R, Chang C, Darden CM, Zurawski S, Boyuk G, Kanak MA, Levy MF, Naziruddin B, Lawrence MC. Pancreatic β-Cell-Derived IP-10/CXCL10 Isletokine Mediates Early Loss of Graft Function in Islet Cell Transplantation. Diabetes 2017; 66:2857-2867. [PMID: 28855240 PMCID: PMC5652609 DOI: 10.2337/db17-0578] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 08/22/2017] [Indexed: 01/08/2023]
Abstract
Pancreatic islets produce and secrete cytokines and chemokines in response to inflammatory and metabolic stress. The physiological role of these "isletokines" in health and disease is largely unknown. We observed that islets release multiple inflammatory mediators in patients undergoing islet transplants within hours of infusion. The proinflammatory cytokine interferon-γ-induced protein 10 (IP-10/CXCL10) was among the highest released, and high levels correlated with poor islet transplant outcomes. Transgenic mouse studies confirmed that donor islet-specific expression of IP-10 contributed to islet inflammation and loss of β-cell function in islet grafts. The effects of islet-derived IP-10 could be blocked by treatment of donor islets and recipient mice with anti-IP-10 neutralizing monoclonal antibody. In vitro studies showed induction of the IP-10 gene was mediated by calcineurin-dependent NFAT signaling in pancreatic β-cells in response to oxidative or inflammatory stress. Sustained association of NFAT and p300 histone acetyltransferase with the IP-10 gene required p38 and c-Jun N-terminal kinase mitogen-activated protein kinase (MAPK) activity, which differentially regulated IP-10 expression and subsequent protein release. Overall, these findings elucidate an NFAT-MAPK signaling paradigm for induction of isletokine expression in β-cells and reveal IP-10 as a primary therapeutic target to prevent β-cell-induced inflammatory loss of graft function after islet cell transplantation.
Collapse
Affiliation(s)
| | | | | | - Rauf Shahbazov
- Islet Cell Laboratory, Baylor Research Institute, Dallas, TX
| | - Charles Chang
- Institute of Biomedical Studies, Baylor University, Waco, TX
| | - Carly M Darden
- Institute of Biomedical Studies, Baylor University, Waco, TX
| | | | - Gulbahar Boyuk
- Adacell Medical Research Center, Department of Endocrinology and Metabolism, Diskapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey
| | - Mazhar A Kanak
- Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Marlon F Levy
- Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Bashoo Naziruddin
- Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, TX
| | | |
Collapse
|
37
|
Stanley WJ, Trivedi PM, Sutherland AP, Thomas HE, Gurzov EN. Differential regulation of pro-inflammatory cytokine signalling by protein tyrosine phosphatases in pancreatic β-cells. J Mol Endocrinol 2017; 59:325-337. [PMID: 28827413 DOI: 10.1530/jme-17-0089] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 08/21/2017] [Indexed: 01/19/2023]
Abstract
Type 1 diabetes (T1D) is characterized by the destruction of insulin-producing β-cells by immune cells in the pancreas. Pro-inflammatory including TNF-α, IFN-γ and IL-1β are released in the islet during the autoimmune assault and signal in β-cells through phosphorylation cascades, resulting in pro-apoptotic gene expression and eventually β-cell death. Protein tyrosine phosphatases (PTPs) are a family of enzymes that regulate phosphorylative signalling and are associated with the development of T1D. Here, we observed expression of PTPN6 and PTPN1 in human islets and islets from non-obese diabetic (NOD) mice. To clarify the role of these PTPs in β-cells/islets, we took advantage of CRISPR/Cas9 technology and pharmacological approaches to inactivate both proteins. We identify PTPN6 as a negative regulator of TNF-α-induced β-cell death, through JNK-dependent BCL-2 protein degradation. In contrast, PTPN1 acts as a positive regulator of IFN-γ-induced STAT1-dependent gene expression, which enhanced autoimmune destruction of β-cells. Importantly, PTPN1 inactivation by pharmacological modulation protects β-cells and primary mouse islets from cytokine-mediated cell death. Thus, our data point to a non-redundant effect of PTP regulation of cytokine signalling in β-cells in autoimmune diabetes.
Collapse
Affiliation(s)
- William J Stanley
- St. Vincent's Institute of Medical ResearchMelbourne, Australia
- Department of MedicineSt. Vincent's Hospital, The University of Melbourne, Melbourne, Australia
| | - Prerak M Trivedi
- St. Vincent's Institute of Medical ResearchMelbourne, Australia
- Department of MedicineSt. Vincent's Hospital, The University of Melbourne, Melbourne, Australia
| | | | - Helen E Thomas
- St. Vincent's Institute of Medical ResearchMelbourne, Australia
- Department of MedicineSt. Vincent's Hospital, The University of Melbourne, Melbourne, Australia
| | - Esteban N Gurzov
- St. Vincent's Institute of Medical ResearchMelbourne, Australia
- Department of MedicineSt. Vincent's Hospital, The University of Melbourne, Melbourne, Australia
- ULB Center for Diabetes ResearchUniversite Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
38
|
Novelli M, Beffy P, Gregorelli A, Porozov S, Mascia F, Vantaggiato C, Masiello P, Menegazzi M. Persistence of STAT-1 inhibition and induction of cytokine resistance in pancreatic β cells treated with St John's wort and its component hyperforin. ACTA ACUST UNITED AC 2017; 71:93-103. [PMID: 28990659 DOI: 10.1111/jphp.12823] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 08/26/2017] [Indexed: 12/22/2022]
Abstract
OBJECTIVES St John's wort extract (SJW) and its component hyperforin (HPF) were shown to potently inhibit cytokine-induced STAT-1 and NF-κB activation in pancreatic β cells and protect them against injury. This study aimed at exploring the time course of STAT-1 inhibition afforded by these natural compounds in the β-cell line INS-1E. METHODS INS-1E cells were pre-incubated with SJW extract (2-5 μg/ml) or HPF (0.5-2 μm) and then exposed to a cytokine mixture. In some experiments, these compounds were added after or removed before cytokine exposure. STAT-1 activation was assessed by electrophoretic mobility shift assay, apoptosis by caspase-3 activity assay, mRNA gene expression by RT-qPCR. KEY FINDINGS Pre-incubation with SJW/HPF for 1-2 h exerted a remarkable STAT-1 downregulation, which was maintained upon removal of the compounds before early or delayed cytokine addition. When the protective compounds were added after cell exposure to cytokines, between 15 and 90 min, STAT-1 inhibition also occurred at a progressively decreasing extent. Upon 24-h incubation, SJW and HPF counteracted cytokine-induced β-cell dysfunction, apoptosis and target gene expression. CONCLUSIONS SJW and HPF confer to β cells a state of 'cytokine resistance', which can be elicited both before and after cytokine exposure and safeguards these cells from deleterious cytokine effects.
Collapse
Affiliation(s)
- Michela Novelli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | - Alex Gregorelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Biochemistry Section, University of Verona, Verona, Italy
| | - Svetlana Porozov
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | - Chiara Vantaggiato
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Pellegrino Masiello
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Marta Menegazzi
- Department of Neurosciences, Biomedicine and Movement Sciences, Biochemistry Section, University of Verona, Verona, Italy
| |
Collapse
|
39
|
Identification of islet-enriched long non-coding RNAs contributing to β-cell failure in type 2 diabetes. Mol Metab 2017; 6:1407-1418. [PMID: 29107288 PMCID: PMC5681241 DOI: 10.1016/j.molmet.2017.08.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 08/10/2017] [Accepted: 08/14/2017] [Indexed: 11/21/2022] Open
Abstract
Objective Non-coding RNAs constitute a major fraction of the β-cell transcriptome. While the involvement of microRNAs is well established, the contribution of long non-coding RNAs (lncRNAs) in the regulation of β-cell functions and in diabetes development remains poorly understood. The aim of this study was to identify novel islet lncRNAs differently expressed in type 2 diabetes models and to investigate their role in β-cell failure and in the development of the disease. Methods Novel transcripts dysregulated in the islets of diet-induced obese mice were identified by high throughput RNA-sequencing coupled with de novo annotation. Changes in the level of the lncRNAs were assessed by real-time PCR. The functional role of the selected lncRNAs was determined by modifying their expression in MIN6 cells and primary islet cells. Results We identified about 1500 novel lncRNAs, a number of which were differentially expressed in obese mice. The expression of two lncRNAs highly enriched in β-cells, βlinc2, and βlinc3, correlated to body weight gain and glycemia levels in obese mice and was also modified in diabetic db/db mice. The expression of both lncRNAs was also modulated in vitro in isolated islet cells by glucolipotoxic conditions. Moreover, the expression of the human orthologue of βlinc3 was altered in the islets of type 2 diabetic patients and was associated to the BMI of the donors. Modulation of the level of βlinc2 and βlinc3 by overexpression or downregulation in MIN6 and mouse islet cells did not affect insulin secretion but increased β-cell apoptosis. Conclusions Taken together, the data show that lncRNAs are modulated in a model of obesity-associated type 2 diabetes and that variations in the expression of some of them may contribute to β-cell failure during the development of the disease.
Mouse pancreatic islets express a large number of novel long non-coding RNAs. Many long non-coding RNAs are differentially expressed in the islets of obese mice. The level of two islet long non-coding RNAs correlates to body weight and glycemia. The expression of these islet long non-coding RNAs is altered in Type 2 diabetes. Altered expression of these long non-coding RNAs sensitise β-cells to apoptosis.
Collapse
|
40
|
Börchers S, Babaei R, Klimpel C, Duque Escobar J, Schröder S, Blume R, Malik MNH, Oetjen E. TNFα-induced DLK activation contributes to apoptosis in the beta-cell line HIT. Naunyn Schmiedebergs Arch Pharmacol 2017; 390:813-825. [DOI: 10.1007/s00210-017-1385-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 05/17/2017] [Indexed: 12/20/2022]
|
41
|
UCP2 Expression Is Increased in Pancreas From Brain-Dead Donors and Involved in Cytokine-Induced β Cells Apoptosis. Transplantation 2017; 101:e59-e67. [DOI: 10.1097/tp.0000000000001292] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
42
|
Villar MMD, Martínez-Abundis E, Preciado-Márquez RO, González-Ortiz M. Effect of diacerein as an add-on to metformin in patients with type 2 diabetes mellitus and inadequate glycemic control. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2017; 61:188-192. [PMID: 28225996 PMCID: PMC10118867 DOI: 10.1590/2359-3997000000242] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 10/10/2016] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To evaluate the effect of diacerein as an add-on to metformin in patients with type 2 diabetes mellitus (T2DM) and inadequate glycemic control. MATERIALS AND METHODS A randomized, double-blind, placebo-controlled clinical trial was carried out on 12 patients with T2DM and inadequate glycemic control [glycated hemoglobin A1c (A1C) ≥ 7%] with metformin as monotherapy (≥ 1500 mg per day) for at least the previous 90 days. Fasting and postprandial glucose were measured before and after the pharmacological intervention. A1C, lipid profile, creatinine and uric acid were also evaluated. After randomization, all patients continued with their dose of metformin. Six subjects received placebo and the other six volunteers took diacerein. Data were tested using the Wilcoxon signed-rank, Mann-Whitney U and chi-square tests. The Institutional Ethics Committee approved the study protocol. RESULTS After 90 days of diacerein as an add-on to metformin, there was a significant decrease in fasting glucose (196 ± 79 vs. 149 ± 70 mg/dL, p < 0.05), postprandial glucose (262 ± 99 vs. 187 ± 70 mg/dlL, p < 0.05) and A1C (8.4 ± 2.0 vs. 6.7 ± 1.7 %, p < 0.05). CONCLUSIONS Diacerein as an add-on to metformin in patients with T2DM improved their glycemic control.
Collapse
|
43
|
Inflammation and the Metabolic Syndrome: The Tissue-Specific Functions of NF-κB. Trends Cell Biol 2017; 27:417-429. [PMID: 28237661 DOI: 10.1016/j.tcb.2017.01.006] [Citation(s) in RCA: 219] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/28/2017] [Accepted: 01/30/2017] [Indexed: 12/16/2022]
Abstract
Obesity is becoming a major health concern in Western society, and medical conditions associated with obesity are grouped in the metabolic syndrome. Overnutrition activates several proinflammatory signaling pathways, leading to a condition of chronic low-grade inflammation in several metabolic tissues affecting their proper function. Nuclear factor kappa B (NF-κB) signaling is a crucial pathway in this process and has been studied extensively in the context of obesity and the metabolic syndrome. Here we give an overview of the molecular mechanisms behind the inflammatory function of NF-κB in response to overnutrition and the effect this has on several metabolic tissues.
Collapse
|
44
|
N-Acetyl-chitobiose ameliorates metabolism dysfunction through Erk/p38 MAPK and histone H3 phosphorylation in type 2 diabetes mice. J Funct Foods 2017. [DOI: 10.1016/j.jff.2016.11.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
45
|
St. John’s wort extract and hyperforin inhibit multiple phosphorylation steps of cytokine signaling and prevent inflammatory and apoptotic gene induction in pancreatic β cells. Int J Biochem Cell Biol 2016; 81:92-104. [DOI: 10.1016/j.biocel.2016.10.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/20/2016] [Accepted: 10/21/2016] [Indexed: 11/20/2022]
|
46
|
Aromatic malononitriles stimulate the resistance of insulin-producing beta-cells to oxidants and inflammatory cytokines. Eur J Pharmacol 2016; 784:69-80. [DOI: 10.1016/j.ejphar.2016.05.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 04/29/2016] [Accepted: 05/09/2016] [Indexed: 01/01/2023]
|
47
|
Cheng L, Zhang D, Chen B. Tumor necrosis factor α-induced protein-3 protects zinc transporter 8 against proinflammatory cytokine-induced downregulation. Exp Ther Med 2016; 12:1509-1514. [PMID: 27588072 DOI: 10.3892/etm.2016.3457] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 04/15/2016] [Indexed: 12/12/2022] Open
Abstract
Zinc transporter 8 (ZnT8) is exclusively expressed in the pancreatic islet and is essential for insulin crystallization, hexamerization and secretion. Tumor necrosis factor α-induced protein-3 (TNFAIP3) is a zinc finger protein that serves a major role in the negative feedback regulation of NF-κB signaling in response to multiple stimuli, and is a central regulator of immunopathology. Although the role of TNFAIP3 in diabetes has been extensively studied, its effect on ZnT8 has not been fully elucidated. The present study aimed to verify whether proinflammatory cytokines, tumor necrosis factor α (TNF-α) and interleukin-1β (IL-1β), are able to affect ZnT8 expression in islet cells. In addition, the study aimed to determine the effect of TNFAIP3 overexpression on cytokine-altered ZnT8 activity, considering its effect on NF-κB signaling. Cell-based studies using NIT-1 cells overexpressing TNFAIP3 were used to assess the effect of cytokines on ZnT8 and NF-κB activation, as well as the effect of TNFAIP3 on ZnT8 expression. Western blot analysis and immunofluorescence staining were employed to determine the protein expression and NF-κB activation, respectively. The results indicated that cytokine stimulation led to TNFAIP3 upregulation, ZnT8 downregulation and NF-κB activation. Furthermore, TNFAIP3 overexpression protected ZnT8 from cytokine-induced downregulation. In conclusion, the current results suggest that inflammation or TNFAIP3 dysfunction may be involved in the pathogenesis of diabetes via ZnT8 expression, besides from islet cell apoptosis. In addition, restricting inflammation and enhancing TNFAIP3 expression may exert a positive effect in diabetes prevention, treatment and pancreatic cell transplantation.
Collapse
Affiliation(s)
- Liqing Cheng
- Department of Endocrinology and Metabolism, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Dongmei Zhang
- Department of Dermatology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Bing Chen
- Department of Endocrinology and Metabolism, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| |
Collapse
|
48
|
Tennant BR, Vanderkruk B, Dhillon J, Dai D, Verchere CB, Hoffman BG. Myt3 suppression sensitizes islet cells to high glucose-induced cell death via Bim induction. Cell Death Dis 2016; 7:e2233. [PMID: 27195679 PMCID: PMC4917670 DOI: 10.1038/cddis.2016.141] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 04/11/2016] [Accepted: 04/22/2016] [Indexed: 12/13/2022]
Abstract
Diabetes is a chronic disease that results from the body's inability to properly control circulating blood glucose levels. The loss of glucose homoeostasis can arise from a loss of β-cell mass because of immune-cell-mediated attack, as in type 1 diabetes, and/or from dysfunction of individual β-cells (in conjunction with target organ insulin resistance), as in type 2 diabetes. A better understanding of the transcriptional pathways regulating islet-cell survival is of great importance for the development of therapeutic strategies that target β-cells for diabetes. To this end, we previously identified the transcription factor Myt3 as a pro-survival factor in islets following acute suppression of Myt3 in vitro. To determine the effects of Myt3 suppression on islet-cell survival in vivo, we used an adenovirus to express an shRNA targeting Myt3 in syngeneic optimal and marginal mass islet transplants, and demonstrate that suppression of Myt3 impairs the function of marginal mass grafts. Analysis of grafts 5 weeks post-transplant revealed that grafts transduced with the shMyt3 adenovirus contained ~20% the number of transduced cells as grafts transduced with a control adenovirus. In fact, increased apoptosis and significant cell loss in the shMyt3-transduced grafts was evident after only 5 days, suggesting that Myt3 suppression sensitizes islet cells to stresses present in the early post-transplant period. Specifically, we find that Myt3 suppression sensitizes islet cells to high glucose-induced cell death via upregulation of the pro-apoptotic Bcl2 family member Bim. Taken together these data suggest that Myt3 may be an important link between glucotoxic and immune signalling pathways.
Collapse
Affiliation(s)
- B R Tennant
- Child and Family Research Institute, British Columbia Children's Hospital, 950 W28th Avenue, Vancouver, British Columbia, Canada V5Z 4H4
| | - B Vanderkruk
- Child and Family Research Institute, British Columbia Children's Hospital, 950 W28th Avenue, Vancouver, British Columbia, Canada V5Z 4H4
| | - J Dhillon
- Child and Family Research Institute, British Columbia Children's Hospital, 950 W28th Avenue, Vancouver, British Columbia, Canada V5Z 4H4
| | - D Dai
- Child and Family Research Institute, British Columbia Children's Hospital, 950 W28th Avenue, Vancouver, British Columbia, Canada V5Z 4H4
| | - C B Verchere
- Child and Family Research Institute, British Columbia Children's Hospital, 950 W28th Avenue, Vancouver, British Columbia, Canada V5Z 4H4.,Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada V5Z 4E3.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada V6T 2B5
| | - B G Hoffman
- Child and Family Research Institute, British Columbia Children's Hospital, 950 W28th Avenue, Vancouver, British Columbia, Canada V5Z 4H4.,Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada V5Z 4E3
| |
Collapse
|
49
|
Brozzi F, Gerlo S, Grieco FA, Juusola M, Balhuizen A, Lievens S, Gysemans C, Bugliani M, Mathieu C, Marchetti P, Tavernier J, Eizirik DL. Ubiquitin D Regulates IRE1α/c-Jun N-terminal Kinase (JNK) Protein-dependent Apoptosis in Pancreatic Beta Cells. J Biol Chem 2016; 291:12040-56. [PMID: 27044747 DOI: 10.1074/jbc.m115.704619] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Indexed: 12/11/2022] Open
Abstract
Pro-inflammatory cytokines contribute to pancreatic beta cell apoptosis in type 1 diabetes at least in part by inducing endoplasmic reticulum (ER) stress and the consequent unfolded protein response (UPR). It remains to be determined what causes the transition from "physiological" to "apoptotic" UPR, but accumulating evidence indicates that signaling by the ER transmembrane protein IRE1α is critical for this transition. IRE1α activation is regulated by both intra-ER and cytosolic cues. We evaluated the role for the presently discovered cytokine-induced and IRE1α-interacting protein ubiquitin D (UBD) on the regulation of IRE1α and its downstream targets. UBD was identified by use of a MAPPIT (mammalian protein-protein interaction trap)-based IRE1α interactome screen followed by comparison against functional genomic analysis of human and rodent beta cells exposed to pro-inflammatory cytokines. Knockdown of UBD in human and rodent beta cells and detailed signal transduction studies indicated that UBD modulates cytokine-induced UPR/IRE1α activation and apoptosis. UBD expression is induced by the pro-inflammatory cytokines interleukin (IL)-1β and interferon (IFN)-γ in rat and human pancreatic beta cells, and it is also up-regulated in beta cells of inflamed islets from non-obese diabetic mice. UBD interacts with IRE1α in human and rodent beta cells, modulating IRE1α-dependent activation of JNK and cytokine-induced apoptosis. Our data suggest that UBD provides a negative feedback on cytokine-induced activation of the IRE1α/JNK pro-apoptotic pathway in cytokine-exposed beta cells.
Collapse
Affiliation(s)
- Flora Brozzi
- From the ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Sarah Gerlo
- the Department of Medical Protein Research, Flanders Interuniversity Institute for Biotechnology (VIB), 9000 Ghent, Belgium, the Department of Biochemistry, Ghent University, 9000 Ghent, Belgium
| | - Fabio Arturo Grieco
- From the ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Matilda Juusola
- From the ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Alexander Balhuizen
- From the ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Sam Lievens
- the Department of Medical Protein Research, Flanders Interuniversity Institute for Biotechnology (VIB), 9000 Ghent, Belgium, the Department of Biochemistry, Ghent University, 9000 Ghent, Belgium
| | - Conny Gysemans
- the Laboratory of Clinical and Experimental Endocrinology, KULeuven, 3000 Leuven, Belgium, and
| | - Marco Bugliani
- the Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, 56126 Pisa, Italy
| | - Chantal Mathieu
- the Laboratory of Clinical and Experimental Endocrinology, KULeuven, 3000 Leuven, Belgium, and
| | - Piero Marchetti
- the Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, 56126 Pisa, Italy
| | - Jan Tavernier
- the Department of Medical Protein Research, Flanders Interuniversity Institute for Biotechnology (VIB), 9000 Ghent, Belgium, the Department of Biochemistry, Ghent University, 9000 Ghent, Belgium
| | - Décio L Eizirik
- From the ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium,
| |
Collapse
|
50
|
Jin R, Xia Y, Chen Q, Li W, Chen D, Ye H, Zhao C, Du X, Shi D, Wu J, Liang G. Da0324, an inhibitor of nuclear factor-κB activation, demonstrates selective antitumor activity on human gastric cancer cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:979-95. [PMID: 27042000 PMCID: PMC4780725 DOI: 10.2147/dddt.s90081] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background The transcription factor nuclear factor-κB (NF-κB) is constitutively activated in a variety of human cancers, including gastric cancer. NF-κB inhibitors that selectively kill cancer cells are urgently needed for cancer treatment. Curcumin is a potent inhibitor of NF-κB activation. Unfortunately, the therapeutic potential of curcumin is limited by its relatively low potency and poor cellular bioavailability. In this study, we presented a novel NF-κB inhibitor named Da0324, a synthetic asymmetric mono-carbonyl analog of curcumin. The purpose of this study is to research the expression of NF-κB in gastric cancer and the antitumor activity and mechanism of Da0324 on human gastric cancer cells. Methods The expressions between gastric cancer tissues/cells and normal gastric tissues/cells of NF-κB were evaluated by Western blot. The inhibition viability of compounds on human gastric cancer cell lines SGC-7901, BGC-823, MGC-803, and normal gastric mucosa epithelial cell line GES-1 was assessed with the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide assay. Absorption spectrum method and high-performance liquid chromatography method detected the stability of the compound in vitro. The compound-induced changes of inducible NF-κB activation in the SGC-7901 and BGC-823 cells were examined by Western blot analysis and immunofluorescence methods. The antitumor activity of compound was performed by clonogenic assay, matrigel invasion assay, flow cytometric analysis, Western blot analysis, and Hoechst 33258 staining assay. Results High levels of p65 were found in gastric cancer tissues and cells. Da0324 displayed higher growth inhibition against several types of gastric cancer cell lines and showed relatively low toxicity to GES-1. Moreover, Da0324 was more stable than curcumin in vitro. Western blot analysis and immunofluorescence methods showed that Da0324 blocked NF-κB activation. In addition, Da0324 significantly inhibited tumor proliferation and invasion, arrested the cell cycle, and induced apoptosis in vitro. Conclusion The asymmetric mono-carbonyl analog of curcumin Da0324 exhibited significantly improved antigastric cancer activity. Da0324 may be a promising NF-κB inhibitor for the selective targeting of cancer cells. However, further studies are needed in animals to validate these findings for the therapeutic use of Da0324.
Collapse
Affiliation(s)
- Rong Jin
- Department of Digestive Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China; Department of Epidemiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Yiqun Xia
- Department of Digestive Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Qiuxiang Chen
- Department of Digestive Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Wulan Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China; College of Information Science and Computer Engineering, Wenzhou Medical College, Wenzhou, People's Republic of China
| | - Dahui Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Hui Ye
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China; School of Basic Medical Sciences, Wenzhou Medical College, Wenzhou, People's Republic of China
| | - Chengguang Zhao
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Xiaojing Du
- Department of Digestive Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Dengjian Shi
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Jianzhang Wu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| |
Collapse
|