1
|
Gurgul-Convey E. To Be or Not to Be: The Divergent Action and Metabolism of Sphingosine-1 Phosphate in Pancreatic Beta-Cells in Response to Cytokines and Fatty Acids. Int J Mol Sci 2022; 23:ijms23031638. [PMID: 35163559 PMCID: PMC8835924 DOI: 10.3390/ijms23031638] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 01/02/2023] Open
Abstract
Sphingosine-1 phosphate (S1P) is a bioactive sphingolipid with multiple functions conveyed by the activation of cell surface receptors and/or intracellular mediators. A growing body of evidence indicates its important role in pancreatic insulin-secreting beta-cells that are necessary for maintenance of glucose homeostasis. The dysfunction and/or death of beta-cells lead to diabetes development. Diabetes is a serious public health burden with incidence growing rapidly in recent decades. The two major types of diabetes are the autoimmune-mediated type 1 diabetes (T1DM) and the metabolic stress-related type 2 diabetes (T2DM). Despite many differences in the development, both types of diabetes are characterized by chronic hyperglycemia and inflammation. The inflammatory component of diabetes remains under-characterized. Recent years have brought new insights into the possible mechanism involved in the increased inflammatory response, suggesting that environmental factors such as a westernized diet may participate in this process. Dietary lipids, particularly palmitate, are substrates for the biosynthesis of bioactive sphingolipids. Disturbed serum sphingolipid profiles were observed in both T1DM and T2DM patients. Many polymorphisms were identified in genes encoding enzymes of the sphingolipid pathway, including sphingosine kinase 2 (SK2), the S1P generating enzyme which is highly expressed in beta-cells. Proinflammatory cytokines and free fatty acids have been shown to modulate the expression and activity of S1P-generating and S1P-catabolizing enzymes. In this review, the similarities and differences in the action of extracellular and intracellular S1P in beta-cells exposed to cytokines or free fatty acids will be identified and the outlook for future research will be discussed.
Collapse
Affiliation(s)
- Ewa Gurgul-Convey
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
2
|
Lei L, Bai G, Wang X, Liu S, Xia J, Wu S, Huan Y, Shen Z. Histone deacetylase 3-selective inhibitor RGFP966 ameliorates impaired glucose tolerance through β-cell protection. Toxicol Appl Pharmacol 2020; 406:115189. [PMID: 32800772 DOI: 10.1016/j.taap.2020.115189] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 08/04/2020] [Accepted: 08/11/2020] [Indexed: 01/18/2023]
Abstract
The potential therapeutic effect of histone deacetylase 3 (HDAC3) pharmacologic inhibition on diabetes has been focused recently. RGFP966, as a highly-selective HDAC3 inhibitor, its possible roles and underlying mechanism in the treatment of diabetes needs to be clarified. In this study, low-dose streptozotocin (STZ)-induced pre-diabetic mice were used to test the regulatory ability of RGFP966 in blood glucose and insulin. We isolated the islets both from normal C57BL/6 J mice and KKAy mice with spontaneous type 2 diabetes to determine the potency of RGFP966 on glucose-stimulated insulin secretion. NIT-1 pancreatic β-cells induced by sodium palmitate (PA) were applied to identify the protective effects of RGFP966 against β-cell apoptosis. The results showed that administration of RGFP966 in the pre-diabetic mice not only significantly reduced hyperglycemia, promoted phase I insulin secretion, improved morphology of islets, but also increased glucose infusion rate (GIR) during hyperglycemic clamp test. When treated in vitro, RGFP966 enhanced insulin secretion and synthesis in islets of normal C57BL/6J mice and diabetic KKAy mice. In addition, it partially attenuated PA-induced apoptosis in NIT-1 cells. Therefore, our research suggests that RGFP966, probably through selective inhibition of HDAC3, might serve as a novel potential preventive and therapeutic candidate for diabetes.
Collapse
Affiliation(s)
- Lei Lei
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Guoliang Bai
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xing Wang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shuainan Liu
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jie Xia
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Song Wu
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yi Huan
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Zhufang Shen
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
3
|
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
|
4
|
Ye C, Driver JP. Suppressors of Cytokine Signaling in Sickness and in Health of Pancreatic β-Cells. Front Immunol 2016; 7:169. [PMID: 27242781 PMCID: PMC4860527 DOI: 10.3389/fimmu.2016.00169] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 04/18/2016] [Indexed: 01/07/2023] Open
Abstract
Suppressors of cytokine signaling (SOCS) are a family of eight proteins that negatively regulate Janus kinase and signal transducers and activators of transcription signaling in cells that utilize this pathway to respond to extracellular stimuli. SOCS are best known for attenuating cytokine signaling in the immune system. However, they are also expressed in many other cell types, including pancreatic β-cells, where there is considerable interest in harnessing SOCS molecules to prevent cytokine-mediated apoptosis during diabetes and allogeneic transplantation. Apart from their potential as therapeutic targets, SOCS molecules play a central role for regulating important functions in β-cells, including growth, glucose sensing, and insulin secretion. This review will discuss SOCS proteins as central regulators for diverse cellular processes important for normal β-cell function as well as their protective anti-apoptotic effects during β-cell stress.
Collapse
Affiliation(s)
- Cheng Ye
- Department of Animal Sciences, University of Florida , Gainesville, FL , USA
| | - John P Driver
- Department of Animal Sciences, University of Florida , Gainesville, FL , USA
| |
Collapse
|
5
|
Diana J, Lehuen A. Macrophages and β-cells are responsible for CXCR2-mediated neutrophil infiltration of the pancreas during autoimmune diabetes. EMBO Mol Med 2015; 6:1090-104. [PMID: 24968718 PMCID: PMC4154135 DOI: 10.15252/emmm.201404144] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Autoimmune type 1 diabetes (T1D) development results from the interaction between pancreatic β-cells, and the innate and the adaptive immune systems culminating with the destruction of the insulin-secreting β-cells by autoreactive T cells. This diabetogenic course starts during the first postnatal weeks by the infiltration of the pancreatic islets by innate immune cells and particularly neutrophils. Here, we aim to determine the cellular and molecular mechanism leading to the recruitment of this neutrophils in the pancreatic islets of non-obese diabetic (NOD) mice. Here, we show that neutrophil recruitment in the pancreatic islets is controlled by inflammatory macrophages and β-cells themselves. Macrophages and β-cells produce the chemokines CXCL1 and CXCL2, recruiting CXCR2-expressing neutrophils from the blood to the pancreatic islets. We further show that pancreatic macrophages secrete IL-1β-inducing CXCR2 ligand production by the β-cells. Finally, the blockade of neutrophil recruitment at early ages using CXCR2 antagonist dampens the diabetogenic T-cell response and the later development of autoimmune diabetes, supporting the therapeutic potential of this approach. Subject Categories Immunology; Metabolism
Collapse
Affiliation(s)
- Julien Diana
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1151, Necker-Enfants Malades Institute (INEM) Necker Hospital, Paris, France Sorbonne Paris Cité, Université Paris Descartes, Paris, France
| | - Agnès Lehuen
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France Institut National de la Santé et de la Recherche Médicale (INSERM), U1016, Cochin Institute Cochin Hospital, Paris, France Laboratoire d'Excellence INFLAMEX, Paris, France
| |
Collapse
|
6
|
Feng X, Tang H, Leng J, Jiang Q. Suppressors of cytokine signaling (SOCS) and type 2 diabetes. Mol Biol Rep 2014; 41:2265-74. [PMID: 24414000 DOI: 10.1007/s11033-014-3079-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 01/04/2014] [Indexed: 12/11/2022]
Abstract
The suppressors of cytokine signaling (SOCS) proteins are originally identified as negative regulators of cytokine-activated Janus kinase/signal transducers and activators of transcription signaling pathway, but increasing evidence reveals that SOCS proteins play an important role in the development of type 2 diabetes involving regulation of the insulin signaling and pancreatic β-cell function, and that SOCS are promising to be the targets for the treatment of type 2 diabetes. In this review, we focus on the emerging role for SOCS and the potential drugs targeting SOCS for type 2 diabetes.
Collapse
Affiliation(s)
- Xiaotao Feng
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, 530001, People's Republic of China
| | | | | | | |
Collapse
|
7
|
The role of chemokines in acute and chronic hepatitis C infection. Cell Mol Immunol 2013; 11:25-40. [PMID: 23954947 DOI: 10.1038/cmi.2013.37] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 07/08/2013] [Accepted: 07/14/2013] [Indexed: 12/12/2022] Open
Abstract
Hepatitis C imposes a significant burden on global healthcare. Chronic infection is associated with progressive inflammation of the liver which typically manifests in cirrhosis, organ failure and cancer. By virtue of elaborate evasion strategies, hepatitis C virus (HCV) succeeds as a persistent human virus. It has an extraordinary capacity to subvert the immune response enabling it to establish chronic infections and associated liver disease. Chemokines are low molecular weight chemotactic peptides that mediate the recruitment of inflammatory cells into tissues and back into the lymphatics and peripheral blood. Thus, they are central to the temporal and spatial distribution of effector and regulatory immune cells. The interactions between chemokines and their cognate receptors help shape the immune response and therefore, have a major influence on the outcome of infection. However, chemokines represent a target for modulation by viruses including the HCV. HCV is known to modulate chemokine expression in vitro and may therefore enable its survival by subverting the immune response in vivo through altered leukocyte chemotaxis resulting in impaired viral clearance and the establishment of chronic low-grade inflammation. In this review, the roles of chemokines in acute and chronic HCV infection are described with a particular emphasis placed on chemokine modulation as a means of immune subversion. We provide an in depth discussion of the part played by chemokines in mediating hepatic fibrosis while addressing the potential applications for these chemoattractants in prognostic medicine.
Collapse
|
8
|
van den Brand BT, Abdollahi-Roodsaz S, Vermeij EA, Bennink MB, Arntz OJ, Rothlin CV, van den Berg WB, van de Loo FAJ. Therapeutic efficacy of Tyro3, Axl, and Mer tyrosine kinase agonists in collagen-induced arthritis. ACTA ACUST UNITED AC 2013. [PMID: 23203851 DOI: 10.1002/art.37786] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Hyperactivation of innate immunity by Toll-like receptors (TLRs) can contribute to the development of autoinflammatory or autoimmune diseases. This study evaluated the activation of Tyro3, Axl, Mer (TAM) receptors, physiologic negative regulators of TLRs, by their agonists, growth arrest-specific protein 6 (GAS-6) and protein S, in the prevention of collagen-induced arthritis (CIA). METHODS Adenoviruses overexpressing GAS-6 and protein S were injected intravenously or intraarticularly into mice during CIA. Splenic T helper cell subsets from intravenously injected mice were studied by flow cytometry, and the knee joints of mice injected intravenously and intraarticularly were assessed histologically. Synovium from mice injected intraarticularly was evaluated for cytokine and suppressor of cytokine signaling (SOCS) expression. RESULTS Protein S significantly reduced ankle joint swelling when overexpressed systemically. Further analysis of knee joints revealed a moderate reduction in pathologic changes in the joint and a significant reduction in the number of splenic Th1 cells when protein S was overexpressed systemically. Local overexpression of GAS-6 decreased joint inflammation and joint pathology. Protein S treatment showed a similar trend of protection. Consistently, GAS-6 and protein S reduced cytokine production in the synovium. Moreover, levels of messenger RNA for interleukin-12 (IL-12) and IL-23 were reduced by GAS-6 and protein S treatment, with a corresponding decrease in the production of interferon-γ and IL-17. TAM ligand overexpression was associated with an increase in SOCS-3 levels, which likely contributed to the amelioration of arthritis. CONCLUSION This study provides the first evidence that TAM receptor stimulation by GAS-6 and protein S can be used to ameliorate arthritis when applied systemically or locally. TAM receptor stimulation limits proinflammatory signaling and adaptive immunity. This pathway provides a novel strategy by which to combat rheumatoid arthritis.
Collapse
Affiliation(s)
- B T van den Brand
- Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
9
|
He L, Wong CK, Cheung KK, Yau HC, Fu A, Zhao HL, Leung KM, Kong AP, Wong GW, Chan PK, Xu G, Chan JC. Anti-inflammatory effects of exendin-4, a glucagon-like peptide-1 analog, on human peripheral lymphocytes in patients with type 2 diabetes. J Diabetes Investig 2013; 4:382-92. [PMID: 24843684 PMCID: PMC4020234 DOI: 10.1111/jdi.12063] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 11/08/2012] [Accepted: 01/07/2013] [Indexed: 02/02/2023] Open
Abstract
Aims/Introduction Type 2 diabetes is characterized by dysregulation of immunity, oxidative stress and reduced incretin effects. Experimental studies suggest that glucagon‐like peptide (GLP‐1) might have immunomodulating effects. We hypothesize that GLP‐1 receptor agonist, exendin‐4, might reduce inflammatory response in type 2 diabetes. Materials and Methods Using peripheral blood mononuclear cells (PBMC) sampled from 10 type 2 diabetes and 10 sex‐ and age‐matched control subjects and supernatants from PBMC culture, the expression of phospho‐mitogen activated protein kinase (MAPK) signaling pathways in CD4+ T helper lymphocytes and monocytes was analyzed using flow cytometry. Cytokines/chemokines and superoxide anion before and after treatment with exendin‐4 were measured by cytometric bead array and chemiluminesence assay, respectively. Results Compared with control subjects, PBMC from type 2 diabetes patients showed activated MAPK (P38, c‐Jun NH2‐terminal protein kinase and extracellular signal‐regulated kinase) signaling pathway, elevated superoxide anion, increased pro‐inflammatory cytokines (tumor necrosis factor‐α, interleukin‐1β, interleukin‐6) and chemokines (CCL5/regulated on activation normal T‐cell expressed and secreted and CXCL10/interferon‐γ‐induced protein 10). These changes were attenuated by exendin‐4, possibly through the suppression of p38 MAPK. Conclusions These results suggest that exendin‐4 might downregulate pro‐inflammatory responses and reduce oxidative stress by suppressing MAPK signaling pathways in type 2 diabetes.
Collapse
Affiliation(s)
- Lan He
- Department of Medicine and Therapeutics The Chinese University of Hong Kong Shatin Hong Kong SAR China
| | - Chun Kwok Wong
- Department of Chemical Pathology The Chinese University of Hong Kong Shatin Hong Kong SAR China
| | - Kitty Kt Cheung
- Department of Medicine and Therapeutics The Chinese University of Hong Kong Shatin Hong Kong SAR China
| | - Ho Chung Yau
- Department of Pediatrics Prince of Wales Hospital The Chinese University of Hong Kong Shatin Hong Kong SAR China
| | - Anthony Fu
- Department of Pediatrics Prince of Wales Hospital The Chinese University of Hong Kong Shatin Hong Kong SAR China
| | - Hai-Lu Zhao
- Department of Medicine and Therapeutics The Chinese University of Hong Kong Shatin Hong Kong SAR China
| | - Karen Ml Leung
- Department of Chemical Pathology The Chinese University of Hong Kong Shatin Hong Kong SAR China
| | - Alice Ps Kong
- Department of Medicine and Therapeutics The Chinese University of Hong Kong Shatin Hong Kong SAR China ; Hong Kong Institute of Diabetes and Obesity The Chinese University of Hong Kong Shatin Hong Kong SAR China ; Li Ka Shing Institute of Health Sciences The Chinese University of Hong Kong Shatin Hong Kong SAR China
| | - Gary Wk Wong
- Department of Pediatrics Prince of Wales Hospital The Chinese University of Hong Kong Shatin Hong Kong SAR China
| | - Paul Ks Chan
- Department of Microbiology The Chinese University of Hong Kong Shatin Hong Kong SAR China
| | - Gang Xu
- Department of Medicine and Therapeutics The Chinese University of Hong Kong Shatin Hong Kong SAR China ; Hong Kong Institute of Diabetes and Obesity The Chinese University of Hong Kong Shatin Hong Kong SAR China ; Li Ka Shing Institute of Health Sciences The Chinese University of Hong Kong Shatin Hong Kong SAR China
| | - Juliana Cn Chan
- Department of Medicine and Therapeutics The Chinese University of Hong Kong Shatin Hong Kong SAR China ; Hong Kong Institute of Diabetes and Obesity The Chinese University of Hong Kong Shatin Hong Kong SAR China ; Li Ka Shing Institute of Health Sciences The Chinese University of Hong Kong Shatin Hong Kong SAR China
| |
Collapse
|
10
|
SOCS and diabetes-ups and downs of a turbulent relationship. Cell Biochem Funct 2013; 31:181-95. [DOI: 10.1002/cbf.2940] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 11/10/2012] [Accepted: 11/16/2012] [Indexed: 11/07/2022]
|
11
|
Wu H, Gao M, Ha T, Kelley J, Young A, Breuel K. Prunella vulgaris aqueous extract attenuates IL-1β-induced apoptosis and NF-κB activation in INS-1 cells. Exp Ther Med 2012; 3:919-924. [PMID: 22969993 DOI: 10.3892/etm.2012.524] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 03/06/2012] [Indexed: 12/13/2022] Open
Abstract
We previously reported that Prunella vulgaris aqueous extract (PVAE) promotes hepatic glycogen synthesis and decreases postprandial hyperglycemia in ICR mice. Inflammatory cytokines play a critical role in the pathogenesis of diabetes. This study was designed to examine whether PVAE has a protective effect on IL-1β-induced apoptosis in INS-1 cells. INS-1 pancreatic β cells were plated at 2×10(6)/ml and treated with PVAE (100 µg/ml) 30 min before the cells were challenged with IL-1β (10 ng/ml). Untreated INS-1 cells served as control. INS-1 cell cytotoxicity was examined by MTT and lactate dehydrogenase (LDH) activity assays. Caspase-3 activity and activation of the apoptotic signaling pathway were analyzed by western blotting. NF-κB binding activity was examined by EMSA. The levels of inflammatory cytokines in the supernatant were measured by ELISA. IL-1β treatment significantly induced INS-1 cell death by 49.2%, increased LDH activity by 1.5-fold and caspase-3 activity by 7.6-fold, respectively, compared with control cells. However, PVAE administration significantly prevented IL-1β-increased INS-1 cell death and LDH activity and attenuated IL-1β-increased caspase-3 activity. Western blot data showed that PVAE also significantly attenuated IL-1β-increased Fas, FasL and phospho-JNK levels in the INS-1 cells. In addition, PVAE treatment significantly attenuated IL-1β-increased NF-κB binding activity and prevented IL-1β-increased TNF-α and IL-6 expression in INS-1 cells. Our data suggest that PVAE has a protective effect on IL-1β-induced INS-1 cell apoptosis. PVAE also attenuates IL-1β-increased NF-κB binding activity and inflammatory cytokine expression in INS-1 cells. PVAE may have a benefit for type I diabetic patients.
Collapse
Affiliation(s)
- Huiping Wu
- Department of Biochemistry, Pre-Clinical College, Nanjing University of Chinese Medicine, Nanjing 210029, P.R. China
| | | | | | | | | | | |
Collapse
|
12
|
Coppola A, Tomasello L, Pizzolanti G, Pucci-Minafra I, Albanese N, Di Cara G, Cancemi P, Pitrone M, Bommarito A, Carissimi E, Zito G, Criscimanna A, Galluzzo A, Giordano C. In vitro phenotypic, genomic and proteomic characterization of a cytokine-resistant murine β-TC3 cell line. PLoS One 2012; 7:e32109. [PMID: 22393382 PMCID: PMC3290556 DOI: 10.1371/journal.pone.0032109] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Accepted: 01/23/2012] [Indexed: 11/19/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is caused by the selective destruction of insulin-producing β-cells. This process is mediated by cells of the immune system through release of nitric oxide, free radicals and pro-inflammatory cytokines, which induce a complex network of intracellular signalling cascades, eventually affecting the expression of genes involved in β-cell survival. The aim of our study was to investigate possible mechanisms of resistance to cytokine-induced β-cell death. To this purpose, we created a cytokine-resistant β-cell line (β-TC3R) by chronically treating the β-TC3 murine insulinoma cell line with IL-1β + IFN-γ. β-TC3R cells exhibited higher proliferation rate and resistance to cytokine-mediated cell death in comparison to the parental line. Interestingly, they maintained expression of β-cell specific markers, such as PDX1, NKX6.1, GLUT2 and insulin. The analysis of the secretory function showed that β-TC3R cells have impaired glucose-induced c-peptide release, which however was only moderately reduced after incubation with KCl and tolbutamide. Gene expression analysis showed that β-TC3R cells were characterized by downregulation of IL-1β and IFN-γ receptors and upregulation of SOCS3, the classical negative regulator of cytokines signaling. Comparative proteomic analysis showed specific upregulation of 35 proteins, mainly involved in cell death, stress response and folding. Among them, SUMO4, a negative feedback regulator in NF-kB and JAK/STAT signaling pathways, resulted hyper-expressed. Silencing of SUMO4 was able to restore sensitivity to cytokine-induced cell death in β-TC3R cells, suggesting it may play a key role in acquired cytokine resistance by blocking JAK/STAT and NF-kB lethal signaling. In conclusion, our study represents the first extensive proteomic characterization of a murine cytokine-resistant β-cell line, which might represent a useful tool for studying the mechanisms involved in resistance to cytokine-mediated β-cell death. This knowledge may be of potential benefit for patients with T1DM. In particular, SUMO4 could be used as a therapeutical target.
Collapse
Affiliation(s)
- Antonina Coppola
- Section of Endocrinology, Dipartimento Biomedico di Medicina Interna e Specialistica (DIBIMIS), University of Palermo, Palermo, Italy
| | - Laura Tomasello
- Section of Endocrinology, Dipartimento Biomedico di Medicina Interna e Specialistica (DIBIMIS), University of Palermo, Palermo, Italy
| | - Giuseppe Pizzolanti
- Section of Endocrinology, Dipartimento Biomedico di Medicina Interna e Specialistica (DIBIMIS), University of Palermo, Palermo, Italy
| | - Ida Pucci-Minafra
- Centro di Oncobiologia Sperimentale (COBS), University of Palermo, Palermo, Italy
| | - Nadia Albanese
- Department of Physics, Centro di Oncobiologia Sperimentale (COBS), University of Palermo, Palermo, Italy
| | - Gianluca Di Cara
- Centro di Oncobiologia Sperimentale (COBS), University of Palermo, Palermo, Italy
| | - Patrizia Cancemi
- Section of Experimental Oncology, Dipartimento Biomedico di Medicina Interna e Specialistica (DIBIMIS), University of Palermo, Palermo, Italy
| | - Maria Pitrone
- Section of Endocrinology, Dipartimento Biomedico di Medicina Interna e Specialistica (DIBIMIS), University of Palermo, Palermo, Italy
| | - Alessandra Bommarito
- Section of Endocrinology, Dipartimento Biomedico di Medicina Interna e Specialistica (DIBIMIS), University of Palermo, Palermo, Italy
| | - Elvira Carissimi
- Section of Endocrinology, Dipartimento Biomedico di Medicina Interna e Specialistica (DIBIMIS), University of Palermo, Palermo, Italy
| | - Giovanni Zito
- Section of Endocrinology, Dipartimento Biomedico di Medicina Interna e Specialistica (DIBIMIS), University of Palermo, Palermo, Italy
| | - Angela Criscimanna
- Section of Endocrinology, Dipartimento Biomedico di Medicina Interna e Specialistica (DIBIMIS), University of Palermo, Palermo, Italy
| | - Aldo Galluzzo
- Section of Endocrinology, Dipartimento Biomedico di Medicina Interna e Specialistica (DIBIMIS), University of Palermo, Palermo, Italy
| | - Carla Giordano
- Section of Endocrinology, Dipartimento Biomedico di Medicina Interna e Specialistica (DIBIMIS), University of Palermo, Palermo, Italy
- Institute of Biomedicine and Molecular Immunology “A. Monroy” (CNR – IBIM), Palermo, Italy
- * E-mail:
| |
Collapse
|
13
|
Börjesson A, Rønn SG, Karlsen AE, Billestrup N, Sandler S. β-cell specific overexpression of suppressor of cytokine signalling-3 does not protect against multiple low dose streptozotocin induced type 1 diabetes in mice. Immunol Lett 2011; 136:74-9. [PMID: 21237203 DOI: 10.1016/j.imlet.2010.12.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2010] [Revised: 10/27/2010] [Accepted: 12/21/2010] [Indexed: 12/21/2022]
Abstract
We investigated the impact of β-cell specific overexpression of suppressor of cytokine signalling-3 (SOCS-3) on the development of multiple low dose streptozotocin (MLDSTZ) induced Type 1 diabetes and the possible mechanisms involved. MLDSTZ treatment was administered to RIP-SOCS-3 transgenic and wild-type (wt) mice and progression of hyperglycemia monitored. Isolated islets from both strains were exposed to human IL-1β (25U/ml) or a combination of human IL-1β (25U/ml) and murine IFN-γ (1000U/ml) for 24h or 48h and we investigated the expression of IL-1 receptor antagonist (IL-1Ra) mRNA in islet cells and secretion of IL-1Ra into culture medium. MLDSTZ treatment caused gradual hyperglycemia both in the wt mice and in the transgenic mice with the latter tending to be more sensitive. In vitro experiments on wt and transgenic islets did not reveal any differences in sensitivity to damaging effects of STZ. Exposure of wt islets to IL-1β or IL-1β+IFN-γ seemed to lead to a failing IL-1Ra response from SOCS-3 transgenic islets. It could be that an increased expression of a possible protective molecule against β-cell destruction may lead to a dampered response of another putative protective molecule. This may have counteracted a protective effect against MLDSTZ in SOCS-3 transgenic mice.
Collapse
Affiliation(s)
- A Börjesson
- Department of Medical Cell Biology, Uppsala University, SE-751 23, Uppsala, Sweden.
| | | | | | | | | |
Collapse
|
14
|
Lebrun P, Cognard E, Gontard P, Bellon-Paul R, Filloux C, Berthault MF, Magnan C, Ruberte J, Luppo M, Pujol A, Pachera N, Herchuelz A, Bosch F, Van Obberghen E. The suppressor of cytokine signalling 2 (SOCS2) is a key repressor of insulin secretion. Diabetologia 2010; 53:1935-46. [PMID: 20499047 DOI: 10.1007/s00125-010-1786-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2010] [Accepted: 04/01/2010] [Indexed: 10/19/2022]
Abstract
AIMS/HYPOTHESIS Suppressor of cytokine signalling (SOCS) proteins are powerful inhibitors of pathways involved in survival and function of pancreatic beta cells. Whereas SOCS1 and SOCS3 have been involved in immune and inflammatory processes, respectively, in beta cells, nothing is known about SOCS2 implication in the pancreas. METHODS Transgenic (tg) mice were generated that constitutively produced SOCS2 in beta cells (betaSOCS2) to define whether this protein is implicated in beta cell functioning and/or survival. RESULTS Constitutive production of SOCS2 in beta cells leads to hyperglycaemia and glucose intolerance. This phenotype is not a consequence of decreased beta cell mass or inhibition of insulin synthesis. However, insulin secretion to various secretagogues is profoundly altered in intact animals and isolated islets. Interestingly, constitutive SOCS2 production dampens the rise in cytosolic free calcium concentration induced by glucose, while glucose metabolism is unchanged. Moreover, tg islets have a depletion in endoplasmic reticulum Ca(2+) stores, suggesting that SOCS2 interferes with calcium fluxes. Finally, in betaSOCS2 mice proinsulin maturation is impaired, leading to an altered structure of insulin secretory granules and augmented levels of proinsulin. The latter is likely to be due to decreased production of prohormone convertase 1 (PC1/3), which plays a key role in proinsulin cleavage. CONCLUSIONS/INTERPRETATIONS SOCS2 was shown to be a potent regulator of proinsulin processing and insulin secretion in beta cells. While its constitutive production is insufficient to induce overt diabetes in this mouse model, it causes glucose intolerance. Thus, increased SOCS2 production could be an important event predisposing to beta cell failure.
Collapse
Affiliation(s)
- P Lebrun
- INSERM U907, Avenue de Valombrose, Nice, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Natalicchio A, De Stefano F, Orlando MR, Melchiorre M, Leonardini A, Cignarelli A, Labarbuta R, Marchetti P, Perrini S, Laviola L, Giorgino F. Exendin-4 prevents c-Jun N-terminal protein kinase activation by tumor necrosis factor-alpha (TNFalpha) and inhibits TNFalpha-induced apoptosis in insulin-secreting cells. Endocrinology 2010; 151:2019-29. [PMID: 20219981 DOI: 10.1210/en.2009-1166] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Glucagon-like peptide-1 and its analogs may preserve pancreatic beta-cell mass by promoting resistance to cytokine-mediated apoptosis. The mechanisms of TNFalpha-induced apoptosis and of its inhibition by exendin-4 were investigated in insulin-secreting cells. INS-1 and MIN6 insulinoma cells were exposed to 20 ng/ml TNFalpha, with or without pretreatment with 10 nm exendin-4. Treatment with TNFalpha increased c-Jun N-terminal protein kinase (JNK) phosphorylation 2-fold, reduced inhibitor-kappaBalpha (IkappaBalpha) protein content by 50%, induced opposite changes in caspase-3 and Bcl-2 protein content, and increased cellular apoptosis. Moreover, exposure to TNFalpha resulted in increased serine phosphorylation of both insulin receptor substrate (IRS)-1 and IRS-2 and reduced basal and insulin-induced Akt phosphorylation. However, in the presence of a JNK inhibitor, TNFalpha-induced apoptosis was diminished and serine phosphorylation of IRS proteins was prevented. When cells were pretreated with exendin-4, TNFalpha-induced JNK and IRS-1/2 serine phosphorylation was markedly reduced, Akt phosphorylation was increased, caspase-3 and Bcl-2 protein levels were restored to normal, and TNFalpha-induced apoptosis was inhibited by 50%. This was associated with a 2-fold increase in IRS-2 expression levels. A similar ability of exendin-4 to prevent TNFalpha-induced JNK phosphorylation was found in isolated pancreatic human islets. The inhibitory effect of exendin-4 on TNFalpha-induced JNK phosphorylation was abrogated in the presence of the protein kinase A inhibitor H89. In conclusion, JNK activation mediates TNFalpha-induced apoptosis and impairment of the IRS/Akt signaling pathway in insulin-secreting cells. By inhibiting JNK phosphorylation in a PKA-dependent manner, exendin-4 counteracts TNFalpha-mediated apoptosis and reverses the inhibitory events in the IRS/Akt pathway, resulting in promotion of cell survival.
Collapse
Affiliation(s)
- Annalisa Natalicchio
- Department of Emergency and Organ Transplantation, Section on Internal Medicine, Endocrinology, Andrology, and Metabolic Diseases, University of Bari, Piazza Giulio Cesare, Bari, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Donath MY, Böni-Schnetzler M, Ellingsgaard H, Halban PA, Ehses JA. Cytokine production by islets in health and diabetes: cellular origin, regulation and function. Trends Endocrinol Metab 2010; 21:261-7. [PMID: 20096598 DOI: 10.1016/j.tem.2009.12.010] [Citation(s) in RCA: 172] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Revised: 12/17/2009] [Accepted: 12/28/2009] [Indexed: 12/21/2022]
Abstract
Islets produce a variety of cytokines and chemokines in response to physiologic and pathologic stimulation by nutrients. The cellular source of these inflammatory mediators includes alpha-, beta-, endothelial-, ductal- and recruited immune cells. Islet-derived cytokines promote alpha- and beta-cell adaptation and repair in the short term. Eventually, chronic metabolic stress can induce a deleterious autoinflammatory process in islets leading to insulin secretion failure and type 2 diabetes. Understanding the specific role of islet derived cytokines and chemokines has opened the door to targeted clinical interventions aimed at remodeling islet inflammation from destruction to adaptation. In this article, we review the islet cellular origin of various cytokines and chemokines and describe their regulation and respective roles in physiology and diabetes.
Collapse
Affiliation(s)
- Marc Y Donath
- Clinic of Endocrinology and Diabetes, Center for Integrated Human Physiology, University Hospital of Zurich, 8091 Zurich, Switzerland.
| | | | | | | | | |
Collapse
|
17
|
Transcriptional regulation of cytokines and oxidative stress by gallic acid in human THP-1 monocytes. Cytokine 2009; 49:229-34. [PMID: 20015662 DOI: 10.1016/j.cyto.2009.11.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Revised: 10/08/2009] [Accepted: 11/05/2009] [Indexed: 11/23/2022]
Abstract
Increased inflammation/prooxidation has been linked not only to Type 2 diabetes but also in prediabetes state. In this study we investigated hyperglycemia-mediated proinflammatory/prooxidant effects in THP-1 monocytes and tested whether gallic acid could attenuate changes in gene expression induced by high-glucose. Cells were treated either with 5.5mM glucose or 25mM glucose in the absence and presence of gallic acid. While oxidative DNA damage was assessed by COMET assay, GSH and GSSG levels were estimated fluorimetrically. Gene expression patterns were determined by RT-PCR. Cells treated with high-glucose showed increased DNA damage and glutathione depletion and this was attenuated in the presence of gallic acid. High-glucose treated cells exhibited increased mRNA expression of TNF-alpha, IL-6, NADPH oxidase and TXNIP and gallic acid attenuated these proinflammatory and prooxidant effects. Cells treated with high-glucose revealed a deficiency in mounting SOCS-3 expression and gallic acid upregulates this feedback regulatory signal. Gallic acid attenuates DNA damage, maintains glutathione turnover, and suppresses hyperglycemia-induced activation of proinflammatory and prooxidant gene expression. Gallic acid beneficially modulate transcription of functionally diverse groups of genes and its regulation of SOCS-3 and TXNIP signals is a newly identified mechanism that has therapeutic implications.
Collapse
|
18
|
Bruun C, Heding PE, Rønn SG, Frobøse H, Rhodes CJ, Mandrup-Poulsen T, Billestrup N. Suppressor of cytokine signalling-3 inhibits Tumor necrosis factor-alpha induced apoptosis and signalling in beta cells. Mol Cell Endocrinol 2009; 311:32-8. [PMID: 19643162 DOI: 10.1016/j.mce.2009.07.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2009] [Revised: 06/19/2009] [Accepted: 07/20/2009] [Indexed: 12/22/2022]
Abstract
Tumor necrosis factor-alpha (TNFalpha) is a pro-inflammatory cytokine involved in the pathogenesis of several diseases including type 1 diabetes mellitus (T1DM). TNFalpha in combination with interleukin-1-beta (IL-1beta) and/or interferon-gamma (IFNgamma) induces specific destruction of the pancreatic insulin-producing beta cells. Suppressor of cytokine signalling-3 (SOCS-3) proteins regulate signalling induced by a number of cytokines including growth hormone, IFNgamma and IL-1beta which signals via very distinctive pathways. The objective of this study was to investigate the effect of SOCS-3 on TNFalpha-induced signalling in beta cells. We found that apoptosis induced by TNFalpha alone or in combination with IL-1beta was suppressed by expression of SOCS-3 in the beta cell line INSr3#2. SOCS-3 inhibited TNFalpha-induced phosphorylation of the mitogen activated protein kinases ERK1/2, p38 and JNK in INSr3#2 cells and in primary rat islets. Furthermore, SOCS-3 repressed TNFalpha-induced degradation of IkappaB, NFkappaB DNA binding and transcription of the NFkappaB-dependent MnSOD promoter. Finally, expression of Socs-3 mRNA was induced by TNFalpha in rat islets in a transient manner with maximum expression after 1-2h. The ability of SOCS-3 to regulate signalling induced by the three major pro-inflammatory cytokines involved in the pathogenesis of T1DM makes SOCS-3 an interesting therapeutic candidate for protection of the beta cell mass.
Collapse
Affiliation(s)
- Christine Bruun
- Hagedorn Research Institute, Niels Steensens Vej 6, NSK2.02, DK-2820 Gentofte, Denmark
| | | | | | | | | | | | | |
Collapse
|
19
|
Ehses JA, Ellingsgaard H, Böni-Schnetzler M, Donath MY. Pancreatic islet inflammation in type 2 diabetes: from alpha and beta cell compensation to dysfunction. Arch Physiol Biochem 2009; 115:240-7. [PMID: 19645635 DOI: 10.1080/13813450903025879] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Evidence in support of the concept of local pancreatic islet inflammation as a mechanism of beta cell failure in type 2 diabetes is accumulating. Observations in human islets from type 2 diabetic patients and rodent models of the disease indicate the increased presence of IL-1 driven cytokines and chemokines in pancreatic islets, concomitant with immune cell infiltration. Inflammation is the body's protective response to harmful stimuli and tissue damage. However, under chronic stress (e.g. metabolic stress in obesity and type 2 diabetes) the body's own defensive response may become deleterious to tissue function. Here, we summarize the current evidence that islet inflammation is a feature of type 2 diabetes, and discuss its role with respect to alpha and beta cell compensation and eventual beta cell failure.
Collapse
Affiliation(s)
- Jan A Ehses
- Division of Endocrinology, Diabetes and Nutrition, Center for Integrated Human Physiology, University Hospital of Zürich, 8091 Zürich, Switzerland.
| | | | | | | |
Collapse
|
20
|
Galicia JC, Benakanakere MR, Stathopoulou PG, Kinane DF. Neutrophils rescue gingival epithelial cells from bacterial-induced apoptosis. J Leukoc Biol 2009; 86:181-6. [PMID: 19389800 DOI: 10.1189/jlb.0109003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In the pathogenesis of chronic inflammatory periodontal disease, neutrophils are recognized as a major cellular component from the histopathology of the periodontal lesion around teeth and from clinical cases where absence or dysfunction of neutrophils results in major periodontal destruction. Neutrophils are recruited in vast numbers into the gingival crevice during periodontal inflammation, attracted by microbial plaque chemoattractants and chemokines released following microbial perturbation of gingival epithelial cells. Porphyromonas gingivalis, a major periodontopathogen, triggers a vast array of cellular responses in gingival epithelial cells but also induces apoptosis. We demonstrate here that neutrophils, when combined in a P. gingivalis challenge assay of epithelial cells, prevent epithelial cell apoptosis by phagocytosing P. gingivalis and later undergoing apoptosis themselves. By removing P. gingivalis by phagocytosis, neutrophils also protect the host from the harmful effects of its microbial proteases, which degrade inflammatory cytokines and other host molecules.
Collapse
Affiliation(s)
- Johnah C Galicia
- University of Louisville School of Dentistry, Louisville, KY 40202, USA
| | | | | | | |
Collapse
|