51
|
Toll-like receptor 4 inhibition prevents autoimmune diabetes in NOD mice. Sci Rep 2019; 9:19350. [PMID: 31852918 PMCID: PMC6920362 DOI: 10.1038/s41598-019-55521-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 11/27/2019] [Indexed: 11/23/2022] Open
Abstract
TLR4 is a transmembrane receptor of the innate immune system that recognizes LPS from gram-negative bacteria. Its stimulation induces pro-inflammatory responses and modulates adaptive immunity. Our aim is to determine the role of TLR4 in the activation and proliferation of T lymphocytes in the onset of autoimmune diabetes, using the non-obese diabetic (NOD) mouse model. Antigen-specific activation and proliferation of diabetogenic T cells were assessed in vitro by Carboxyfluorescein succinimidyl ester (CFSE) dilution, in presence of vehicle or CLI-095, a cyclohexene derivative that inhibits TLR4 signaling. NOD mice were treated with vehicle or CLI-095 and sacrificed either before or after the onset of autoimmune diabetes. T lymphocyte activation and proliferation were evaluated in treated and control mice. Insulitis was analyzed by histology and diabetes incidence was determined in treated and control mice. Our results demonstrate that TLR4 blockade decreases CD4+ T lymphocyte activation and auto-antigen-specific proliferation both in vitro and in vivo, decreases the infiltrative insulitis and finally prevents the onset of spontaneous diabetes. Taken together, our data demonstrate that TLR4 signaling contributes to the development and maintenance of autoimmune diabetes. The immunomodulatory effect of CLI-095 could be part of a preventive strategy targeting patients at risk for type 1 diabetes.
Collapse
|
52
|
Wang Y, Yu H, Liu F, Song X. Analysis of key genes and their functions in placental tissue of patients with gestational diabetes mellitus. Reprod Biol Endocrinol 2019; 17:104. [PMID: 31783860 PMCID: PMC6884804 DOI: 10.1186/s12958-019-0546-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 11/20/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND This study was aimed at screening out the potential key genes and pathways associated with gestational diabetes mellitus (GDM). METHODS The GSE70493 dataset used for this study was obtained from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) in the placental tissue of women with GDM in relation to the control tissue samples were identified and submitted to protein-protein interaction (PPI) network analysis and subnetwork module mining. Functional enrichment analyses of the PPI network and subnetworks were subsequently carried out. Finally, the integrated miRNA-transcription factor (TF)-DEG regulatory network was analyzed. RESULTS In total, 238 DEGs were identified, of which 162 were upregulated and 76 were downregulated. Through PPI network construction, 108 nodes and 278 gene pairs were obtained, from which chemokine (C-X-C motif) ligand 9 (CXCL9), CXCL10, protein tyrosine phosphatase, receptor type C (PTPRC), and human leukocyte antigen (HLA) were screened out as hub genes. Moreover, genes associated with the immune-related pathway and immune responses were found to be significantly enriched in the process of GDM. Finally, miRNAs and TFs that target the DEGs were predicted. CONCLUSIONS Four candidate genes (viz., CXCL9, CXCL10, PTPRC, and HLA) are closely related to GDM. miR-223-3p, miR-520, and thioredoxin-binding protein may play important roles in the pathogenesis of this disease.
Collapse
Affiliation(s)
- Yuxia Wang
- grid.452222.1Department of Gynecology, Jinan Central Hospital, Jinan City, 250013 Shandong Province China
| | - Haifeng Yu
- grid.452222.1Department of Obstetrics, Jinan Central Hospital, No. 105 Jiefang Road, Lixia District, Jinan City, 250013 Shandong Province China
| | - Fangmei Liu
- grid.452222.1Department of Obstetrics, Jinan Central Hospital, No. 105 Jiefang Road, Lixia District, Jinan City, 250013 Shandong Province China
| | - Xiue Song
- grid.452222.1Department of Obstetrics, Jinan Central Hospital, No. 105 Jiefang Road, Lixia District, Jinan City, 250013 Shandong Province China
| |
Collapse
|
53
|
Ardestani A, Li S, Annamalai K, Lupse B, Geravandi S, Dobrowolski A, Yu S, Zhu S, Baguley TD, Surakattula M, Oetjen J, Hauberg-Lotte L, Herranz R, Awal S, Altenhofen D, Nguyen-Tran V, Joseph S, Schultz PG, Chatterjee AK, Rogers N, Tremblay MS, Shen W, Maedler K. Neratinib protects pancreatic beta cells in diabetes. Nat Commun 2019; 10:5015. [PMID: 31676778 PMCID: PMC6825211 DOI: 10.1038/s41467-019-12880-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 10/02/2019] [Indexed: 02/06/2023] Open
Abstract
The loss of functional insulin-producing β-cells is a hallmark of diabetes. Mammalian sterile 20-like kinase 1 (MST1) is a key regulator of pancreatic β-cell death and dysfunction; its deficiency restores functional β-cells and normoglycemia. The identification of MST1 inhibitors represents a promising approach for a β-cell-protective diabetes therapy. Here, we identify neratinib, an FDA-approved drug targeting HER2/EGFR dual kinases, as a potent MST1 inhibitor, which improves β-cell survival under multiple diabetogenic conditions in human islets and INS-1E cells. In a pre-clinical study, neratinib attenuates hyperglycemia and improves β-cell function, survival and β-cell mass in type 1 (streptozotocin) and type 2 (obese Leprdb/db) diabetic mouse models. In summary, neratinib is a previously unrecognized inhibitor of MST1 and represents a potential β-cell-protective drug with proof-of-concept in vitro in human islets and in vivo in rodent models of both type 1 and type 2 diabetes. Type 1 as well as type 2 diabetes are characterized by a loss of insulin-producing β-cells. Here the authors show that the FDA-approved drug neratinib has beneficial effects on β-cell survival, insulin secretion, and glycemic control in mouse models of diabetes.
Collapse
Affiliation(s)
- Amin Ardestani
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany.
| | - Sijia Li
- Calibr at Scripps Research, La Jolla, CA, USA
| | - Karthika Annamalai
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Blaz Lupse
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Shirin Geravandi
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | | | - Shan Yu
- Calibr at Scripps Research, La Jolla, CA, USA
| | - Siying Zhu
- Calibr at Scripps Research, La Jolla, CA, USA
| | | | | | - Janina Oetjen
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany.,Center for Industrial Mathematics, University of Bremen, Bremen, Germany.,MALDI Imaging Lab, University of Bremen, Bremen, Germany
| | - Lena Hauberg-Lotte
- Center for Industrial Mathematics, University of Bremen, Bremen, Germany
| | - Raquel Herranz
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Sushil Awal
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Delsi Altenhofen
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | | | - Sean Joseph
- Calibr at Scripps Research, La Jolla, CA, USA
| | | | | | | | | | - Weijun Shen
- Calibr at Scripps Research, La Jolla, CA, USA.
| | - Kathrin Maedler
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany.
| |
Collapse
|
54
|
Ježek P, Jabůrek M, Plecitá-Hlavatá L. Contribution of Oxidative Stress and Impaired Biogenesis of Pancreatic β-Cells to Type 2 Diabetes. Antioxid Redox Signal 2019; 31:722-751. [PMID: 30450940 PMCID: PMC6708273 DOI: 10.1089/ars.2018.7656] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 11/05/2018] [Indexed: 12/14/2022]
Abstract
Significance: Type 2 diabetes development involves multiple changes in β-cells, related to the oxidative stress and impaired redox signaling, beginning frequently by sustained overfeeding due to the resulting lipotoxicity and glucotoxicity. Uncovering relationships among the dysregulated metabolism, impaired β-cell "well-being," biogenesis, or cross talk with peripheral insulin resistance is required for elucidation of type 2 diabetes etiology. Recent Advances: It has been recognized that the oxidative stress, lipotoxicity, and glucotoxicity cannot be separated from numerous other cell pathology events, such as the attempted compensation of β-cell for the increased insulin demand and dynamics of β-cell biogenesis and its "reversal" at dedifferentiation, that is, from the concomitantly decreasing islet β-cell mass (also due to transdifferentiation) and low-grade islet or systemic inflammation. Critical Issues: At prediabetes, the compensation responses of β-cells, attempting to delay the pathology progression-when exaggerated-set a new state, in which a self-checking redox signaling related to the expression of Ins gene expression is impaired. The resulting altered redox signaling, diminished insulin secretion responses to various secretagogues including glucose, may lead to excretion of cytokines or chemokines by β-cells or excretion of endosomes. They could substantiate putative stress signals to the periphery. Subsequent changes and lasting glucolipotoxicity promote islet inflammatory responses and further pathology spiral. Future Directions: Should bring an understanding of the β-cell self-checking and related redox signaling, including the putative stress signal to periphery. Strategies to cure or prevent type 2 diabetes could be based on the substitution of the "wrong" signal by the "correct" self-checking signal.
Collapse
Affiliation(s)
- Petr Ježek
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martin Jabůrek
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Lydie Plecitá-Hlavatá
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
55
|
Zhang W, Dong X, Wang T, Kong Y. Exosomes derived from platelet-rich plasma mediate hyperglycemia-induced retinal endothelial injury via targeting the TLR4 signaling pathway. Exp Eye Res 2019; 189:107813. [PMID: 31560926 DOI: 10.1016/j.exer.2019.107813] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 09/01/2019] [Accepted: 09/23/2019] [Indexed: 01/07/2023]
Abstract
In this study, we aimed to investigate whether exosomes derived from platelet-rich plasma (PRP-Exos) can regulate hyperglycemia-induced retinal injury via targeting the TLR4 signaling pathway. We studied the effects of PRP-Exos on retinal endothelial injury in diabetic rats and human retinal endothelial cells (HRECs) in vitro. Isolated PRP-Exos were observed by transmission electron microscopy and flow cytometry. Samples were obtained from the retinas of rats and cultured HRECs after treatment to analyze reactive oxygen species levels. Immunofluorescence and Western blotting were conducted to assess the levels of adhesion molecules and the TLR4 signaling pathway. The content of CXCL10 in PRP-Exos was analyzed by Western blot. The plasma level of PRP-Exos was greatly increased in diabetic rats. In cultured HRECs, PRP-Exos induced the production of malonyldialdehyde(MDA) and reactive oxygen species(ROS) and inhibited the activity of superoxide dismutase(SOD). Further analysis showed that the activation of the TLR4 pathway by PRP-Exos played a pivotal role in regulating inflammation. The inhibition of the TLR4 pathway by TAK-242 had a robust protective effect on PRP-Exo-induced retinal endothelial injury in vitro and vivo. In addition, PRP-Exo-derived CXCL10 led to retinal endothelial injury, and antagonizing CXCL10 with a CXCL10-neutralizing antibody dramatically attenuated such injury. In summary, PRP-Exos mediate hyperglycemia-induced retinal endothelial injury by upregulating the TLR4 signaling pathway.
Collapse
Affiliation(s)
- Wei Zhang
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Clinical College of Ophthalmology Tianjin Medical University, Tianjin, 300020, China
| | - Xue Dong
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Tian Wang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yichun Kong
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Clinical College of Ophthalmology Tianjin Medical University, Tianjin, 300020, China.
| |
Collapse
|
56
|
Scharfmann R, Staels W, Albagli O. The supply chain of human pancreatic β cell lines. J Clin Invest 2019; 129:3511-3520. [PMID: 31478912 PMCID: PMC6715382 DOI: 10.1172/jci129484] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Patients with type 1 or type 2 diabetes have an insufficiency in their functional β cell mass. To advance diabetes treatment and to work toward a cure, a better understanding of how to protect the pancreatic β cells against autoimmune or metabolic assaults (e.g., obesity, gestation) will be required. Over the past decades, β cell protection has been extensively investigated in rodents both in vivo and in vitro using isolated islets or rodent β cell lines. Transferring these rodent data to humans has long been challenging, at least partly for technical reasons: primary human islet preparations were scarce and functional human β cell lines were lacking. In 2011, we described a robust protocol of targeted oncogenesis in human fetal pancreas and produced the first functional human β cell line, and in subsequent years additional lines with specific traits. These cell lines are currently used by more than 150 academic and industrial laboratories worldwide. In this Review, we first explain how we developed the human β cell lines and why we think we succeeded where others, despite major efforts, did not. Next, we discuss the use of such functional human β cell lines and share some perspectives on their use to advance diabetes research.
Collapse
Affiliation(s)
- Raphael Scharfmann
- INSERM U1016, Institut Cochin, Université Paris Descartes, Paris, France
| | - Willem Staels
- INSERM U1016, Institut Cochin, Université Paris Descartes, Paris, France
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Brussels, Belgium
| | - Olivier Albagli
- INSERM U1016, Institut Cochin, Université Paris Descartes, Paris, France
| |
Collapse
|
57
|
Mizgier ML, Fernández-Verdejo R, Cherfan J, Pinget M, Bouzakri K, Galgani JE. Insights on the Role of Putative Muscle-Derived Factors on Pancreatic Beta Cell Function. Front Physiol 2019; 10:1024. [PMID: 31440170 PMCID: PMC6694406 DOI: 10.3389/fphys.2019.01024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 07/24/2019] [Indexed: 01/02/2023] Open
Abstract
Skeletal muscle is a main target of insulin action that plays a pivotal role in postprandial glucose disposal. Importantly, skeletal muscle insulin sensitivity relates inversely with pancreatic insulin secretion, which prompted the hypothesis of the existence of a skeletal muscle-pancreas crosstalk mediated through an endocrine factor. The observation that changes in skeletal muscle glucose metabolism are accompanied by altered insulin secretion supports this hypothesis. Meanwhile, a muscle-derived circulating factor affecting in vivo insulin secretion remains elusive. This factor may correspond to peptides/proteins (so called myokines), exosomes and their cargo, and metabolites. We hereby review the most remarkable evidence encouraging the possibility of such inter-organ communication, with special focus on muscle-derived factors that may potentially mediate such skeletal muscle-pancreas crosstalk.
Collapse
Affiliation(s)
- Maria L Mizgier
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Strasbourg, France
| | - Rodrigo Fernández-Verdejo
- Departamento de Ciencias de la Salud, Nutrición y Dietética, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Julien Cherfan
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Strasbourg, France
| | - Michel Pinget
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Strasbourg, France
| | - Karim Bouzakri
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Strasbourg, France
| | - Jose E Galgani
- Departamento de Ciencias de la Salud, Nutrición y Dietética, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Nutrición, Diabetes y Metabolismo, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
58
|
Lee T, Yun S, Jeong JH, Jung TW. Asprosin impairs insulin secretion in response to glucose and viability through TLR4/JNK-mediated inflammation. Mol Cell Endocrinol 2019; 486:96-104. [PMID: 30853600 DOI: 10.1016/j.mce.2019.03.001] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 03/02/2019] [Accepted: 03/04/2019] [Indexed: 01/02/2023]
Abstract
Severe inflammation in the islets is observed in obese patients with type 2 diabetes. Inflammation in the islets is caused by obesity-induced serum free fatty acids. Asprosin is a fasting-induced adipokine, which contributes to hepatic glucose production. However, the effects of asprosin on inflammation and cellular dysfunction in pancreatic β-cells remain to be elucidated. Here, we demonstrated that treatment of mouse insulinoma MIN6 cells and human primary islets containing β-cells with palmitate increased asprosin expression and secretion. Treatment of MIN6 cells and human primary islets with palmitate increased phosphorylation of the inflammatory marker nuclear factor-kappa B (NFκB) and the release of pro-inflammatory cytokines including TNF and MCP-1 and decreased glucose-stimulated insulin secretion and cell viability. However, siRNA-mediated suppression of asprosin reversed these changes. Recombinant asprosin treatment of MIN6 cells and human primary islets augmented the inflammation response, cellular dysfunction, and apoptosis in a dose-dependent manner. Asprosin induced toll-like receptor (TLR) 4 expression and JNK phosphorylation. siRNA for TLR4 or JNK mitigated the effects of asprosin on inflammation and cellular dysfunction. These results suggest that palmitate-derived asprosin secretion from β-cells results in their inflammation and dysfunction through a TLR4/JNK-mediated pathway. This report suggests asprosin as a novel therapeutic target for the treatment of type 2 diabetes through preservation of β-cell function.
Collapse
Affiliation(s)
- Taeseung Lee
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, South Korea; Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Subin Yun
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, South Korea
| | - Tae Woo Jung
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, South Korea.
| |
Collapse
|
59
|
Watson D, Yang JYC, Sarwal RD, Sigdel TK, Liberto JM, Damm I, Louie V, Sigdel S, Livingstone D, Soh K, Chakraborty A, Liang M, Lin PC, Sarwal MM. A Novel Multi-Biomarker Assay for Non-Invasive Quantitative Monitoring of Kidney Injury. J Clin Med 2019; 8:E499. [PMID: 31013714 PMCID: PMC6517941 DOI: 10.3390/jcm8040499] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/01/2019] [Accepted: 04/10/2019] [Indexed: 12/11/2022] Open
Abstract
The current standard of care measures for kidney function, proteinuria, and serum creatinine (SCr) are poor predictors of early-stage kidney disease. Measures that can detect chronic kidney disease in its earlier stages are needed to enable therapeutic intervention and reduce adverse outcomes of chronic kidney disease. We have developed the Kidney Injury Test (KIT) and a novel KIT Score based on the composite measurement and validation of multiple biomarkers across a unique set of 397 urine samples. The test is performed on urine samples that require no processing at the site of collection and without target sequencing or amplification. We sought to verify that the pre-defined KIT test, KIT Score, and clinical thresholds correlate with established chronic kidney disease (CKD) and may provide predictive information on early kidney injury status above and beyond proteinuria and renal function measurements alone. Statistical analyses across six DNA, protein, and metabolite markers were performed on a subset of residual spot urine samples with CKD that met assay performance quality controls from patients attending the clinical labs at the University of California, San Francisco (UCSF) as part of an ongoing IRB-approved prospective study. Inclusion criteria included selection of patients with confirmed CKD and normal healthy controls; exclusion criteria included incomplete or missing information for sample classification, logistical delays in transport/processing of urine samples or low sample volume, and acute kidney injury. Multivariate logistic regression of kidney injury status and likelihood ratio statistics were used to assess the contribution of the KIT Score for prediction of kidney injury status and stage of CKD as well as assess the potential contribution of the KIT Score for detection of early-stage CKD above and beyond traditional measures of renal function. Urine samples were processed by a proprietary immunoprobe for measuring cell-free DNA (cfDNA), methylated cfDNA, clusterin, CXCL10, total protein, and creatinine. The KIT Score and stratified KIT Score Risk Group (high versus low) had a sensitivity and specificity for detection of kidney injury status (healthy or CKD) of 97.3% (95% CI: 94.6-99.3%) and 94.1% (95% CI: 82.3-100%). In addition, in patients with normal renal function (estimated glomerular filtration rate (eGFR) ≥ 90), the KIT Score clearly identifies those with predisposing risk factors for CKD, which could not be detected by eGFR or proteinuria (p < 0.001). The KIT Score uncovers a burden of kidney injury that may yet be incompletely recognized, opening the door for earlier detection, intervention and preservation of renal function.
Collapse
Affiliation(s)
- Drew Watson
- KIT Bio, 665 3rd Street, San Francisco, CA 94107, USA.
| | - Joshua Y C Yang
- KIT Bio, 665 3rd Street, San Francisco, CA 94107, USA.
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
- Masters in Translational Medicine Program, University of California Berkeley, Berkeley, CA 94720, USA.
| | - Reuben D Sarwal
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Tara K Sigdel
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Juliane M Liberto
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Izabella Damm
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Victoria Louie
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Shristi Sigdel
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Devon Livingstone
- Masters in Translational Medicine Program, University of California Berkeley, Berkeley, CA 94720, USA.
| | - Katherine Soh
- Masters in Translational Medicine Program, University of California Berkeley, Berkeley, CA 94720, USA.
| | - Arjun Chakraborty
- Masters in Translational Medicine Program, University of California Berkeley, Berkeley, CA 94720, USA.
| | - Michael Liang
- Masters in Translational Medicine Program, University of California Berkeley, Berkeley, CA 94720, USA.
| | - Pei-Chen Lin
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
- Masters in Translational Medicine Program, University of California Berkeley, Berkeley, CA 94720, USA.
| | - Minnie M Sarwal
- KIT Bio, 665 3rd Street, San Francisco, CA 94107, USA.
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
- Masters in Translational Medicine Program, University of California Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
60
|
Zhou JH, Cai JJ, She ZG, Li HL. Noninvasive evaluation of nonalcoholic fatty liver disease: Current evidence and practice. World J Gastroenterol 2019; 25:1307-1326. [PMID: 30918425 PMCID: PMC6429343 DOI: 10.3748/wjg.v25.i11.1307] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/20/2019] [Accepted: 02/23/2019] [Indexed: 02/06/2023] Open
Abstract
With the increasing number of individuals with diabetes and obesity, nonalcoholic fatty liver disease (NAFLD) is becoming increasingly prevalent, affecting one-quarter of adults worldwide. The spectrum of NAFLD ranges from simple steatosis or nonalcoholic fatty liver (NAFL) to nonalcoholic steatohepatitis (NASH). NAFLD, especially NASH, may progress to fibrosis, leading to cirrhosis and hepatocellular carcinoma. NAFLD can impose a severe economic burden, and patients with NAFLD-related terminal or deteriorative liver diseases have become one of the main groups receiving liver transplantation. The increasing prevalence of NAFLD and the severe outcomes of NASH make it necessary to use effective methods to identify NAFLD. Although recognized as the gold standard, biopsy is limited by its sampling bias, poor acceptability, and severe complications, such as mortality, bleeding, and pain. Therefore, noninvasive methods are urgently needed to avoid biopsy for diagnosing NAFLD. This review discusses the current noninvasive methods for assessing NAFLD, including steatosis, NASH, and NAFLD-related fibrosis, and explores the advantages and disadvantages of measurement tools. In addition, we analyze potential noninvasive biomarkers for tracking disease processes and monitoring treatment effects, and explore effective algorithms consisting of imaging and nonimaging biomarkers for diagnosing advanced fibrosis and reducing unnecessary biopsies in clinical practice.
Collapse
Affiliation(s)
- Jiang-Hua Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Institute of Model Animal of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Jing-Jing Cai
- Department of Cardiology, The 3rd Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Institute of Model Animal of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Hong-Liang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Institute of Model Animal of Wuhan University, Wuhan 430071, Hubei Province, China
| |
Collapse
|
61
|
Xu W, Morford J, Mauvais-Jarvis F. Emerging role of testosterone in pancreatic β-cell function and insulin secretion. J Endocrinol 2019; 240:JOE-18-0573.R1. [PMID: 30601759 PMCID: PMC6602868 DOI: 10.1530/joe-18-0573] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 12/24/2018] [Indexed: 12/16/2022]
Abstract
One of the most sexually dimorphic aspects of metabolic regulation is the bidirectional modulation of glucose homeostasis by testosterone in male and females. Severe testosterone deficiency predisposes men to type 2 diabetes (T2D), while in contrast, androgen excess predisposes women to hyperglycemia. The role of androgen deficiency and excess in promoting visceral obesity and insulin resistance in men and women respectively is well established. However, although it is established that hyperglycemia requires β cell dysfunction to develop, the role of testosterone in β cell function is less understood. This review discusses recent evidence that the androgen receptor (AR) is present in male and female β cells. In males, testosterone action on AR in β cells enhances glucose-stimulated insulin secretion by potentiating the insulinotropic action of glucagon-like peptide-1. In females, excess testosterone action via AR in β cells promotes insulin hypersecretion leading to oxidative injury, which in turn predisposes to T2D.
Collapse
Affiliation(s)
- Weiwei Xu
- W Xu, Division of Endocrinology and Metabolism, Tulane University, New Orleans, United States
| | - Jamie Morford
- J Morford, Division of Endocrinology and Metabolism, Tulane University, New Orleans, United States
| | - Franck Mauvais-Jarvis
- F Mauvais-Jarvis, Division of Endocrinology and Metabolism, Tulane University, New Orleans, United States
| |
Collapse
|
62
|
He W, Rebello O, Savino R, Terracciano R, Schuster-Klein C, Guardiola B, Maedler K. TLR4 triggered complex inflammation in human pancreatic islets. Biochim Biophys Acta Mol Basis Dis 2018; 1865:86-97. [PMID: 30287405 DOI: 10.1016/j.bbadis.2018.09.030] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 09/06/2018] [Accepted: 09/20/2018] [Indexed: 02/08/2023]
Abstract
Type 2 Diabetes (T2D) is strongly associated with obesity and inflammation. Toll-like receptor-4 (TLR-4) is the major pro-inflammatory pathway with its ligands and downstream products increased systemically in T2D and in at-risk individuals. Detailed mechanisms of the complex proinflammatory response in pancreatic islets remain unknown. In isolated human islets LPS induced IL-1β, IL-6, IL-8 and TNF production in a TLR4-dependent manner and severely impaired β-cell survival and function. IL-6 antagonism improved β-cell function. IL-8, which was identified specifically in α-cells, initiated monocyte migration, a process fully blocked by IL-8 neutralization. The TLR4 response was potentiated in obese donors; with higher IL-1β, IL-6 and IL-8 expression than in non-obese donors. TLR4 activation leads to a complex multi-cellular inflammatory response in human islets, which involves β-cell failure, cytokine production and macrophage recruitment to islets. In obesity, the amplified TLR4 response may potentiate β-cell damage and accelerate diabetes progression.
Collapse
Affiliation(s)
- Wei He
- University of Bremen, Center for Biomolecular Interactions Bremen, Germany.
| | - Osmond Rebello
- University of Bremen, Center for Biomolecular Interactions Bremen, Germany
| | - Rocco Savino
- Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
| | - Rosa Terracciano
- Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
| | | | | | - Kathrin Maedler
- University of Bremen, Center for Biomolecular Interactions Bremen, Germany.
| |
Collapse
|
63
|
Early TLR4 Blockade Attenuates Sterile Inflammation-mediated Stress in Islets During Isolation and Promotes Successful Transplant Outcomes. Transplantation 2018; 102:1505-1513. [DOI: 10.1097/tp.0000000000002287] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
64
|
Lebreton F, Berishvili E, Parnaud G, Rouget C, Bosco D, Berney T, Lavallard V. NLRP3 inflammasome is expressed and regulated in human islets. Cell Death Dis 2018; 9:726. [PMID: 29941940 PMCID: PMC6018156 DOI: 10.1038/s41419-018-0764-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 06/05/2018] [Accepted: 06/08/2018] [Indexed: 02/02/2023]
Abstract
NRLP3 inflammasome is a protein complex involved in the maturation of IL1β. In the onset of type 1 diabetes as well as in islet transplantation, IL-1β is one of the cytokines involved in the recruitment of immune cells in islets and eventually in islet destruction. Whether IL-1β is produced by islet cells is still under debate and NLRP3 inflammasome-dependent IL-1β production has not been yet determined in human islets. The aim of this study was to determine the expression and the regulation of the NRLP3 inflammasome in human islets. Human islets were stimulated with LPS and successively with ATP (LPS + ATP) in the presence or absence of the inflammasome inhibitor glyburide. Islets were also incubated in hypoxic or normoxic conditions for 24 h in the presence or absence of glyburide. Then, IL1B and NLRP3 expression was studied by real time PCR, protein expression by western blot, protein localization by immunofluorescence and protein secretion by ELISA. LPS + ATP increased gene expression of NRLP3 and IL1B. Glyburide partially prevented this effect. IL-1β protein was localized in β and non-β cells. Moreover, LPS + ATP increased IL-1β protein expression and production, which were prevented by glyburide. Hypoxia increased gene expression of NRLP3 and IL1B and induced IL-1β and caspase-1 production. Finally, hypoxia-induced cell death which was not prevented by inhibition of NLRP3 inflammasome. NRLP3 inflammasome is expressed and plays a role in IL-1β production by human islets. By contrast, NRLP3 inflammasome activation is not involved in islet cell death induced by hypoxia.
Collapse
Affiliation(s)
- Fanny Lebreton
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University, Hospitals and University of Geneva, Geneva, Switzerland
| | - Ekaterine Berishvili
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University, Hospitals and University of Geneva, Geneva, Switzerland
| | - Géraldine Parnaud
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University, Hospitals and University of Geneva, Geneva, Switzerland
| | - Caroline Rouget
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University, Hospitals and University of Geneva, Geneva, Switzerland
| | - Domenico Bosco
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University, Hospitals and University of Geneva, Geneva, Switzerland
| | - Thierry Berney
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University, Hospitals and University of Geneva, Geneva, Switzerland
| | - Vanessa Lavallard
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University, Hospitals and University of Geneva, Geneva, Switzerland.
| |
Collapse
|
65
|
Hueso L, Ortega R, Selles F, Wu-Xiong NY, Ortega J, Civera M, Ascaso JF, Sanz MJ, Real JT, Piqueras L. Upregulation of angiostatic chemokines IP-10/CXCL10 and I-TAC/CXCL11 in human obesity and their implication for adipose tissue angiogenesis. Int J Obes (Lond) 2018; 42:1406-1417. [PMID: 29795466 DOI: 10.1038/s41366-018-0102-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/04/2018] [Accepted: 04/10/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND/AIMS Impaired angiogenesis is linked to adipose tissue (AT) dysfunction, inflammation, and insulin resistance in human obesity. Chemokine (C-X-C motif) receptor. (CXCR3) ligands are important regulators of angiogenesis in different disease contexts such as cancer; however, their role in human morbid obesity is unknown. We investigated the role of the CXCR3 axis in AT angiogenesis in morbidly obese patients. SUBJECTS/METHODS The study group comprised 50 morbidly obese patients (mean age 44 ± 1 years, body mass index 44 ± 1 kg/m2) who had undergone laparoscopic Roux-Y-gastric bypass surgery, and 25 age-matched non-obese control subjects. We measured the circulating levels of the CXCR3 ligands monokine induced by interferon-γ (MIG/CXCL9), interferon-γ inducible protein 10 (IP-10/CXCL10), and interferon-γ-inducible T-cell alpha chemoattractant (I-TAC/CXCL11) in all studied subjects. Additionally, the expression of CXCR3 ligands was analyzed in paired biopsies of subcutaneous and visceral AT obtained during the laparoscopic procedure in morbidly obese patients. Additionally, we explored the functional role of CXCR3 ligands on angiogenesis in AT from morbidly obese patients using an ex vivo assay. RESULTS Plasma levels of CXCL10 and CXCL11 were significantly higher in morbidly obese patients than in controls (p < 0.01). In ex vivo assays, angiogenic growth was markedly lower in visceral AT than in subcutaneous AT (p < 0.05), which was related to significant tissue upregulation of CXCL10, CXCL11 and CXCR3 (p < 0.05). CXCL10 or CXCL11 inhibited AT angiogenesis (p < 0.05), and blockade of CXCR3 function significantly increased capillary sprouting in visceral fat deposits (p < 0.05). Western blot analysis showed that the p38 mitogen-activated protein kinase signaling pathway was implicated in the angiostatic effects of CXCR3 in AT. CONCLUSIONS CXCL10 and CXCL11 may play. deleterious role in obesity as potential inhibitors of AT angiogenesis. Accordingly, pharmacological blockade of CXCR3 could represent. therapy to prevent AT dysfunction in obesity.
Collapse
Affiliation(s)
- Luisa Hueso
- Institute of Health Research-INCLIVA, Valencia, Spain
| | - Rebeca Ortega
- Institute of Health Research-INCLIVA, Valencia, Spain.,Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | | | - Ning Yun Wu-Xiong
- Institute of Health Research-INCLIVA, Valencia, Spain.,Endocrinology and Nutrition Service, University Clinic Hospital of Valencia, Valencia, Spain
| | - Joaquin Ortega
- Surgery Service, University Clinic Hospital of Valencia, Valencia, Spain.,Department of Surgery, University of Valencia, Valencia, Spain
| | - Miguel Civera
- Institute of Health Research-INCLIVA, Valencia, Spain.,Endocrinology and Nutrition Service, University Clinic Hospital of Valencia, Valencia, Spain
| | - Juan F Ascaso
- Institute of Health Research-INCLIVA, Valencia, Spain.,Endocrinology and Nutrition Service, University Clinic Hospital of Valencia, Valencia, Spain.,CIBERDEM: Diabetes and Associated Metabolic Diseases Networking Biomedical Research- ISCIII, Madrid, Spain
| | - Maria-Jesus Sanz
- Institute of Health Research-INCLIVA, Valencia, Spain.,Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - José T Real
- Institute of Health Research-INCLIVA, Valencia, Spain. .,Endocrinology and Nutrition Service, University Clinic Hospital of Valencia, Valencia, Spain. .,CIBERDEM: Diabetes and Associated Metabolic Diseases Networking Biomedical Research- ISCIII, Madrid, Spain.
| | - Laura Piqueras
- Institute of Health Research-INCLIVA, Valencia, Spain. .,Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain.
| |
Collapse
|
66
|
Xu C, Zhang C, Ji J, Wang C, Yang J, Geng B, Zhao T, Zhou H, Mu X, Pan J, Hu S, Lv Y, Chen X, Wen H, You Q. CD36 deficiency attenuates immune-mediated hepatitis in mice by modulating the proapoptotic effects of CXC chemokine ligand 10. Hepatology 2018; 67:1943-1955. [PMID: 29220536 DOI: 10.1002/hep.29716] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 10/30/2017] [Accepted: 12/02/2017] [Indexed: 12/21/2022]
Abstract
UNLABELLED The scavenger receptor CD36 recognizes a diverse set of ligands and has been implicated in a wide variety of normal and pathological processes, including lipid metabolism, angiogenesis, atherosclerosis, and phagocytosis. In particular, recent findings have demonstrated its crucial functions in sterile inflammation and tumor metastasis. However, the role of CD36 in immune-mediated hepatitis remains unclear. Concanavalin A (ConA)-induced liver injury is a well-established experimental T cell-mediated hepatitis. To understand the role of CD36 in hepatitis, we tested the susceptibility of CD36-deficient (CD36-/- ) mice to this model, evaluated by a liver enzyme test, terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assay, histological analysis, mononuclear cell (MNC) infiltration, and hepatic proinflammatory factor production. CD36-/- mice were less sensitive to ConA-induced hepatitis and had a significantly lower number of liver MNCs (LMNCs), including CD4+ cells, CD8+ T cells, natural killer cells, natural killer T cells, infiltrating macrophages, and neutrophils, as well as reduced expression of inflammatory mediators (tumor necrosis factor α, CXC chemokine ligand (CXCL) 10, interleukin (IL)-1α, monocyte chemotactic protein 1, and IL-6) compared with controls. Notably, we used bone marrow chimeric mice to demonstrate that CD36 expression on nonhematopoietic cells was required to drive ConA-induced liver injury. Furthermore, our data show that the CD36 receptor was essential for CXCL10-induced hepatocyte apoptosis and activation of IκB kinase, Akt, and Jun N-terminal kinase. Moreover, treatment of wild-type mice with genistein, a tyrosine kinase inhibitor that blocks CD36-Lyn signaling, attenuated ConA-induced liver injury and reduced the number of MNCs. CONCLUSIONS Our findings suggest that CD36 plays an important proinflammatory role in ConA-induced liver injury by promoting hepatic inflammation and mediating the proapoptotic effect of chemokine CXCL10, and therefore, may be a potential therapeutic target for immune-mediated hepatitis. (Hepatology 2018;67:1943-1955).
Collapse
Affiliation(s)
- Che Xu
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chen Zhang
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jie Ji
- First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chao Wang
- Department of Surgery, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jing Yang
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Biao Geng
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ting Zhao
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hong Zhou
- Department of Immunology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xianmin Mu
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jinshun Pan
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shi Hu
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuanfang Lv
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xingguo Chen
- Department of Rheumatology and Immunology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hao Wen
- Department of Surgery, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qiang You
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Immunology, Nanjing Medical University, Nanjing, Jiangsu, China
- Medical Center for Digestive Diseases, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
67
|
Barlow JP, Solomon TP. Do skeletal muscle-secreted factors influence the function of pancreatic β-cells? Am J Physiol Endocrinol Metab 2018; 314:E297-E307. [PMID: 29208613 DOI: 10.1152/ajpendo.00353.2017] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Skeletal muscle is an endocrine organ that secretes a variety of compounds including proteins (myokines), metabolites, microRNAs (miRNAs), and exosomes, many of which are regulated by exercise and play important roles in endocrine signaling. Interorgan communication via muscle-secreted factors therefore provides a novel area for investigation and implicates the importance of skeletal muscle in the pathophysiology of metabolic diseases such as type 2 diabetes (T2D). Given that underlying molecular mechanisms of T2D are subject of ongoing research, in light of new evidence it is probable that interorgan cross-talk between skeletal muscle and pancreatic β-cells plays an important part. To date, the number of studies published in this field provide the basis of this review. Specifically, we discuss current experimental evidence in support for a role of skeletal muscle to β-cell cross-talk, paying particular attention to muscle-secreted factors including myokines, metabolites, miRNAs, and factors contained within exosomes that influence the function and/or the survival of β-cells in health and disease. In reviewing this evidence, we provide an update on the list of known muscle-secreted factors that have potential to influence the function and/or survival of β-cells under normal and diabetic conditions. We also report limitations of current cross-talk methods and discuss future directions in this growing field.
Collapse
Affiliation(s)
- Jonathan P Barlow
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham , Birmingham, West Midlands , United Kingdom
| | - Thomas P Solomon
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham , Birmingham, West Midlands , United Kingdom
| |
Collapse
|
68
|
An SCF FBXO28 E3 Ligase Protects Pancreatic β-Cells from Apoptosis. Int J Mol Sci 2018; 19:ijms19040975. [PMID: 29587369 PMCID: PMC5979299 DOI: 10.3390/ijms19040975] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 01/09/2023] Open
Abstract
Loss of pancreatic β-cell function and/or mass is a central hallmark of all forms of diabetes but its molecular basis is incompletely understood. β-cell apoptosis contributes to the reduced β-cell mass in diabetes. Therefore, the identification of important signaling molecules that promote β-cell survival in diabetes could lead to a promising therapeutic intervention to block β-cell decline during development and progression of diabetes. In the present study, we identified F-box protein 28 (FBXO28), a substrate-recruiting component of the Skp1-Cul1-F-box (SCF) ligase complex, as a regulator of pancreatic β-cell survival. FBXO28 was down-regulated in β-cells and in isolated human islets under diabetic conditions. Consistently, genetic silencing of FBXO28 impaired β-cell survival, and restoration of FBXO28 protected β-cells from the harmful effects of the diabetic milieu. Although FBXO28 expression positively correlated with β-cell transcription factor NEUROD1 and FBXO28 depletion also reduced insulin mRNA expression, neither FBXO28 overexpression nor depletion had any significant impact on insulin content, glucose-stimulated insulin secretion (GSIS) or on other genes involved in glucose sensing and metabolism or on important β-cell transcription factors in isolated human islets. Consistently, FBXO28 overexpression did not further alter insulin content and GSIS in freshly isolated islets from patients with type 2 diabetes (T2D). Our data show that FBXO28 improves pancreatic β-cell survival under diabetogenic conditions without affecting insulin secretion, and its restoration may be a novel therapeutic tool to promote β-cell survival in diabetes.
Collapse
|
69
|
Azizi Z, Lange C, Paroni F, Ardestani A, Meyer A, Wu Y, Zander AR, Westenfelder C, Maedler K. β-MSCs: successful fusion of MSCs with β-cells results in a β-cell like phenotype. Oncotarget 2018; 7:48963-48977. [PMID: 27374092 PMCID: PMC5226484 DOI: 10.18632/oncotarget.10214] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 04/19/2016] [Indexed: 12/22/2022] Open
Abstract
Bone marrow mesenchymal stromal cells (MSC) have anti-inflammatory, anti-apoptotic and immunosuppressive properties and are a potent source for cell therapy. Cell fusion has been proposed for rapid generation of functional new reprogrammed cells. In this study, we aimed to establish a fusion protocol of bone marrow−derived human MSCs with the rat beta-cell line (INS-1E) as well as human isolated pancreatic islets in order to generate insulin producing beta-MSCs as a cell-based treatment for diabetes. Human eGFP+ puromycin+ MSCs were co-cultured with either stably mCherry-expressing rat INS-1E cells or human dispersed islet cells and treated with phytohemagglutinin (PHA-P) and polyethylene glycol (PEG) to induce fusion. MSCs and fused cells were selected by puromycin treatment. With an improved fusion protocol, 29.8 ± 2.9% of all MSCs were β-MSC heterokaryons based on double positivity for mCherry and eGFP. After fusion and puromycin selection, human NKX6.1 and insulin as well as rat Neurod1, Nkx2.2, MafA, Pdx1 and Ins1 mRNA were highly elevated in fused human MSC/INS-1E cells, compared to the mixed control population. Such induction of beta-cell markers was confirmed in fused human MSC/human dispersed islet cells, which showed elevated NEUROD1, NKX2.2, MAFA, PDX1 and insulin mRNA compared to the mixed control. Fused cells had higher insulin content and improved insulin secretion compared to the mixed control and insulin positive beta-MSCs also expressed nuclear PDX1. We established a protocol for fusion of human MSCs and beta cells, which resulted in a beta cell like phenotype. This could be a novel tool for cell-based therapies of diabetes.
Collapse
Affiliation(s)
- Zahra Azizi
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany.,Department of Cell and Gene Therapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claudia Lange
- Department of Cell and Gene Therapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Federico Paroni
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany
| | - Amin Ardestani
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany
| | - Anke Meyer
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany
| | - Yonghua Wu
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany.,Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Axel R Zander
- Department of Cell and Gene Therapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christof Westenfelder
- Departments of Medicine and Physiology, University of Utah and George E Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Kathrin Maedler
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany.,German Center for Diabetes Research (DZD) project partner, University of Bremen, Bremen, Germany
| |
Collapse
|
70
|
He W, Yuan T, Choezom D, Hunkler H, Annamalai K, Lupse B, Maedler K. Ageing potentiates diet-induced glucose intolerance, β-cell failure and tissue inflammation through TLR4. Sci Rep 2018; 8:2767. [PMID: 29426925 PMCID: PMC5807311 DOI: 10.1038/s41598-018-20909-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/25/2018] [Indexed: 12/25/2022] Open
Abstract
Ageing and obesity are two major risk factors for the development of type 2 diabetes (T2D). A chronic, low-grade, sterile inflammation contributes to insulin resistance and β-cell failure. Toll-like receptor-4 (TLR4) is a major pro-inflammatory pathway; its ligands as well as downstream signals are increased systemically in patients with T2D and at-risk individuals. In the present study we investigated the combined effects of high fat/high sucrose diet (HFD) feeding, ageing and TLR4-deficiency on tissue inflammation, insulin resistance and β-cell failure. In young mice, a short-term HFD resulted in a mildly impaired glucose tolerance and reduced insulin secretion, together with a β-cell mass compensation. In older mice, HFD further deteriorated insulin secretion and induced a significantly impaired glucose tolerance and augmented tissue inflammation in adipose, liver and pancreatic islets, all of which was attenuated by TLR4 deficiency. Our results show that ageing exacerbates HFD-induced impairment of glucose homeostasis and pancreatic β-cell function and survival, and deteriorates HFD-induced induction of mRNA expression of inflammatory cytokines and pro-inflammatory macrophage markers. TLR4-deficiency protects against these combined deleterious effects of a high fat diet and ageing through a reduced expression of inflammatory products in both insulin sensitive tissues and pancreatic islets.
Collapse
Affiliation(s)
- Wei He
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany.
| | - Ting Yuan
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany
| | - Dolma Choezom
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany
| | - Hannah Hunkler
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany
| | - Karthika Annamalai
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany
| | - Blaz Lupse
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany
| | - Kathrin Maedler
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany.
| |
Collapse
|
71
|
Dodington DW, Desai HR, Woo M. JAK/STAT - Emerging Players in Metabolism. Trends Endocrinol Metab 2018; 29:55-65. [PMID: 29191719 DOI: 10.1016/j.tem.2017.11.001] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/06/2017] [Accepted: 11/06/2017] [Indexed: 02/07/2023]
Abstract
The Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway is crucial for transducing signals from a variety of metabolically relevant hormones and cytokines including growth hormone, leptin, erythropoietin, IL4, IL6 and IFNγ. A growing body of evidence suggests that this pathway is dysregulated in the context of obesity and metabolic disease. Recent development of animal models has been instrumental in identifying the role of JAK/STAT signaling in the peripheral metabolic organs including adipose, liver, muscle, pancreas, and the immune system. In this review we summarize current knowledge about the function of JAK/STAT proteins in the regulation of metabolism, and highlight new potential therapeutic targets for the treatment of obesity and diabetes.
Collapse
Affiliation(s)
- David W Dodington
- Toronto General Hospital Research Institute, University Health Network, Toronto, M5G 1L7, Canada
| | - Harsh R Desai
- Toronto General Hospital Research Institute, University Health Network, Toronto, M5G 1L7, Canada
| | - Minna Woo
- Toronto General Hospital Research Institute, University Health Network, Toronto, M5G 1L7, Canada; Division of Endocrinology and Metabolism, Department of Medicine, University Health Network and University of Toronto, Toronto, M5G 2C4, Canada.
| |
Collapse
|
72
|
Chrysin Induces Antidiabetic, Antidyslipidemic and Anti-Inflammatory Effects in Athymic Nude Diabetic Mice. Molecules 2017; 23:molecules23010067. [PMID: 29283418 PMCID: PMC6017955 DOI: 10.3390/molecules23010067] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 12/24/2017] [Accepted: 12/27/2017] [Indexed: 01/22/2023] Open
Abstract
Extensive knowledge of diabetes and its complications is helpful to find new drugs for proper treatment to stop degenerative changes derived from this disease. In this context, chrysin (5,7-dihydroxyflavone) is a natural product that occurs in a variety of flowers and fruits with anti-inflammatory and antidiabetic effects, among others. Thus, a diabetic model in athymic nude mice was developed and used to establish the ability of chrysin to decrease the secretion of pro-inflammatory cytokines. Also, it was determined the acute (50 mg/kg) and sub-acute (50 mg/kg/day/10 days) antidiabetic and antihyperlipidemic activities after the period of time treatment. Results indicate that chrysin has significant acute antihyperglycemic and antidiabetic effects in nude diabetic mice (p < 0.05). Moreover, triglyceride blood levels were reduced and IL-1β and TNF-α were diminished after 10 days’ treatment compared with control group (p < 0.05). In conclusion, it was found that chrysin could produce similar effects as metformin, a drug used for the treatment of diabetes, since both test samples decreased glucose and triglycerides levels, they impaired the generation of pro-inflammatory cytokines involved in the development of diabetes and its consequences, such as atherosclerosis and other cardiovascular diseases.
Collapse
|
73
|
Lee JH, Kim B, Jin WJ, Kim HH, Ha H, Lee ZH. Pathogenic roles of CXCL10 signaling through CXCR3 and TLR4 in macrophages and T cells: relevance for arthritis. Arthritis Res Ther 2017; 19:163. [PMID: 28724396 PMCID: PMC5518115 DOI: 10.1186/s13075-017-1353-6] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 05/30/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by uncontrolled joint inflammation and destruction of bone and cartilage. We previously reported that C-X-C motif chemokine 10 (CXCL10; also called IP-10) has important roles in joint inflammation and bone destruction in arthritis. However, the specific mechanisms by which CXCL10 regulates the recruitment of inflammatory cells and the production of osteoclastogenic cytokines in RA progression are not fully understood. METHODS Bone marrow-derived macrophages and CD4+ T cells were isolated from wild-type (WT), Cxcl10 -/-, and Cxcr3 -/- mice. CXCL10-induced migration was performed using a Boyden chamber, and CXCL10-stimulated production of osteoclastogenic cytokines was measured by quantitative real-time PCR and ELISA. Collagen antibody-induced arthritis (CAIA) was induced by administration of collagen type II antibodies and lipopolysaccharide to the mice. Clinical scores were analyzed and hind paws were collected for high-resolution micro-CT, and histomorphometry. Serum was used to assess bone turnover and levels of osteoclastogenic cytokines. RESULTS CXCL10 increased the migration of inflammatory cells through C-X-C chemokine receptor 3 (CXCR3)-mediated, but not toll-like receptor 4 (TLR4)-mediated, ERK activation. Interestingly, both receptors CXCR3 and TLR4 were simultaneously required for CXCL10-stimulated production of osteoclastogenic cytokines in CD4+ T cells. Furthermore, calcineurin-dependent NFATc1 activation was essential for CXCL10-induced RANKL expression. In vivo, F4/80+ macrophages and CD4+ T cells robustly infiltrated into synovium of WT mice with CAIA but were significantly reduced in both Cxcl10 -/- and Cxcr3 -/- mice. Serum concentrations of osteoclastogenic cytokines and bone destruction were also reduced in the knockout mice, leading to attenuated progression of arthritis. CONCLUSION These findings highlight the importance of CXCL10 signaling in the pathogenesis of RA and provide previously unidentified details of the mechanisms by which CXCL10 promotes the development of arthritis.
Collapse
Affiliation(s)
- Jong-Ho Lee
- Brain Tumor Center and Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Bongjun Kim
- Department of Cell and Developmental Biology, Dental Research Institute, School of Dentistry, Seoul National University, 28 Yeongon-dong, Jongno-gu, Seoul, 110-749, Republic of Korea
| | - Won Jong Jin
- Department of Cell and Developmental Biology, Dental Research Institute, School of Dentistry, Seoul National University, 28 Yeongon-dong, Jongno-gu, Seoul, 110-749, Republic of Korea
| | - Hong-Hee Kim
- Department of Cell and Developmental Biology, Dental Research Institute, School of Dentistry, Seoul National University, 28 Yeongon-dong, Jongno-gu, Seoul, 110-749, Republic of Korea
| | - Hyunil Ha
- Clinical Research Division, Korean Medicine-Based Herbal Drug Development Group, Korea Institute of Oriental Medicine, 483 Expo-Ro, Yuseong-Gu, Daejeon, 305-811, Republic of Korea.
| | - Zang Hee Lee
- Department of Cell and Developmental Biology, Dental Research Institute, School of Dentistry, Seoul National University, 28 Yeongon-dong, Jongno-gu, Seoul, 110-749, Republic of Korea.
| |
Collapse
|
74
|
Schludi B, Moin ASM, Montemurro C, Gurlo T, Matveyenko AV, Kirakossian D, Dawson DW, Dry SM, Butler PC, Butler AE. Islet inflammation and ductal proliferation may be linked to increased pancreatitis risk in type 2 diabetes. JCI Insight 2017; 2:92282. [PMID: 28679961 DOI: 10.1172/jci.insight.92282] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 05/23/2017] [Indexed: 12/26/2022] Open
Abstract
Pancreatitis is more frequent in type 2 diabetes mellitus (T2DM), although the underlying cause is unknown. We tested the hypothesis that ongoing β cell stress and apoptosis in T2DM induces ductal tree proliferation, particularly the pancreatic duct gland (PDG) compartment, and thus potentially obstructs exocrine outflow, a well-established cause of pancreatitis. PDG replication was increased 2-fold in human pancreas from individuals with T2DM, and was associated with increased pancreatic intraepithelial neoplasia (PanIN), lesions associated with pancreatic inflammation and with the potential to obstruct pancreatic outflow. Increased PDG replication in the prediabetic human-IAPP-transgenic (HIP) rat model of T2DM was concordant with increased β cell stress but preceded metabolic derangement. Moreover, the most abundantly expressed chemokines released by the islets in response to β cell stress in T2DM, CXCL1, -4, and -10, induced proliferation in human pancreatic ductal epithelium. Also, the diabetes medications reported as potential modifiers for the risk of pancreatitis in T2DM modulated PDG proliferation accordingly. We conclude that chronic stimulation and proliferation of the PDG compartment in response to islet inflammation in T2DM is a potentially novel mechanism that serves as a link to the increased risk for pancreatitis in T2DM and may potentially be modified by currently available diabetes therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - David W Dawson
- Department of Pathology and Laboratory Medicine.,Jonsson Comprehensive Cancer Center, UCLA, David Geffen School of Medicine, Los Angeles, California, USA
| | - Sarah M Dry
- Department of Pathology and Laboratory Medicine.,Jonsson Comprehensive Cancer Center, UCLA, David Geffen School of Medicine, Los Angeles, California, USA
| | - Peter C Butler
- Larry L. Hillblom Islet Research Center.,Jonsson Comprehensive Cancer Center, UCLA, David Geffen School of Medicine, Los Angeles, California, USA
| | | |
Collapse
|
75
|
Collier JJ, Sparer TE, Karlstad MD, Burke SJ. Pancreatic islet inflammation: an emerging role for chemokines. J Mol Endocrinol 2017; 59:R33-R46. [PMID: 28420714 PMCID: PMC5505180 DOI: 10.1530/jme-17-0042] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 04/18/2017] [Indexed: 12/13/2022]
Abstract
Both type 1 and type 2 diabetes exhibit features of inflammation associated with alterations in pancreatic islet function and mass. These immunological disruptions, if unresolved, contribute to the overall pathogenesis of disease onset. This review presents the emerging role of pancreatic islet chemokine production as a critical factor regulating immune cell entry into pancreatic tissue as well as an important facilitator of changes in tissue resident leukocyte activity. Signaling through two specific chemokine receptors (i.e., CXCR2 and CXCR3) is presented to illustrate key points regarding ligand-mediated regulation of innate and adaptive immune cell responses. The prospective roles of chemokine ligands and their corresponding chemokine receptors to influence the onset and progression of autoimmune- and obesity-associated forms of diabetes are discussed.
Collapse
MESH Headings
- Adaptive Immunity
- Animals
- Chemokines/genetics
- Chemokines/immunology
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/immunology
- Diabetes Mellitus, Type 2/pathology
- Disease Models, Animal
- Gene Expression Regulation
- Humans
- Immunity, Innate
- Inflammation
- Islets of Langerhans/immunology
- Islets of Langerhans/pathology
- Leukocytes/immunology
- Leukocytes/pathology
- Obesity/genetics
- Obesity/immunology
- Obesity/pathology
- Receptors, CXCR3/genetics
- Receptors, CXCR3/immunology
- Receptors, Interleukin-8B/genetics
- Receptors, Interleukin-8B/immunology
- Signal Transduction
Collapse
Affiliation(s)
- J Jason Collier
- Laboratory of Islet Biology and InflammationPennington Biomedical Research Center, Baton Rouge, Louisiana, USA
- Department of SurgeryGraduate School of Medicine, University of Tennessee Health Science Center, Knoxville, Tennessee, USA
| | - Tim E Sparer
- Department of MicrobiologyUniversity of Tennessee, Knoxville, Knoxville, Tennessee, USA
| | - Michael D Karlstad
- Department of SurgeryGraduate School of Medicine, University of Tennessee Health Science Center, Knoxville, Tennessee, USA
| | - Susan J Burke
- Laboratory of ImmunogeneticsPennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| |
Collapse
|
76
|
Deng H, Li Z, Liu G, Li X, Chen Y, Zhang Y, Sun Y, Fu J. Elevated serum interferon γ-inducible protein-10 in women with polycystic ovary syndrome. Gynecol Endocrinol 2017; 33:363-367. [PMID: 28051885 DOI: 10.1080/09513590.2016.1269740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Interferon γ-induced protein 10 kDa (IP10/CXCL10) is a chemokine related to endocrine disorders; however, the serum concentrations of IP10 in women with polycystic ovary syndrome (PCOS) have not yet been reported. Therefore, we investigated whether IP10 is increased in PCOS patients and its potential clinical value in PCOS patients. METHODS For this research, the serum IP10, glucose, insulin, high sensitivity C-reactive protein (hs-CRP), follicle-stimulating hormone (FSH), luteinizing hormone (LH) and total testosterone (TT) concentrations were measured in 60 women with PCOS and healthy controls. RESULTS The median IP10 concentration was 45.60 pg/mL [interquartile range (IQR):29.75, 79.69], which was significantly higher than that of the body mass index (BMI)-matched controls (median: 36.46 pg/mL; IQR:28.98, 45.80). In the multivariate linear regression analysis, hs-CRP and the homeostasis model assessment of insulin resistance index (HOMA2-IR) were independent predictors of the IP10 values, while FSH was inversely associated with the IP10.No significant association was observed between the IP10 and BMI, glucose, LH and TT. CONCLUSIONS The serum IP10 concentrations increase in women with PCOS, moreover, IP10 appears to be correlated with the inflammatory and IR statuses of PCOS. IP10 may be a potential biomarker to estimate the disease activity of PCOS.
Collapse
Affiliation(s)
- Hongli Deng
- a Department of Clinical Laboratory , Affiliated Liuyang Hospital of University of South China, People's Hospital of Liuyang City , Changsha , Hunan , China
| | - Zhibo Li
- a Department of Clinical Laboratory , Affiliated Liuyang Hospital of University of South China, People's Hospital of Liuyang City , Changsha , Hunan , China
| | - Guang Liu
- a Department of Clinical Laboratory , Affiliated Liuyang Hospital of University of South China, People's Hospital of Liuyang City , Changsha , Hunan , China
| | - Xianhua Li
- a Department of Clinical Laboratory , Affiliated Liuyang Hospital of University of South China, People's Hospital of Liuyang City , Changsha , Hunan , China
| | - Yong Chen
- b Department of Clinical Laboratory , Affiliated Changsha Hospital of University of South China, The First Hospital of Changsha City , Changsha , Hunan , China
| | - Yong Zhang
- c Department of Gastrointestinal Surgery , Affiliated Liuyang Hospital of University of South China, People's Hospital of Liuyang City , Chang sha , Hunan , China
| | - Yifan Sun
- d Department of Clinical Laboratory , Third Affiliated Hospital of Guangxi University of Chinese Medicine , Liuzhou , Guangxi , China , and
| | - Jinjian Fu
- e Department of Clinical Laboratory , Liuzhou Maternity and Child Health Care Hospital , Liuzhou , Guangxi , China
| |
Collapse
|
77
|
Xu W, Niu T, Xu B, Navarro G, Schipma MJ, Mauvais-Jarvis F. Androgen receptor-deficient islet β-cells exhibit alteration in genetic markers of insulin secretion and inflammation. A transcriptome analysis in the male mouse. J Diabetes Complications 2017; 31:787-795. [PMID: 28343791 PMCID: PMC5472375 DOI: 10.1016/j.jdiacomp.2017.03.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/06/2017] [Accepted: 03/06/2017] [Indexed: 11/19/2022]
Abstract
AIMS Testosterone action is mediated via the androgen receptor (AR). We have reported that male mice lacking AR selectively in β-cells (βARKO-/y) develop decreased glucose-stimulated insulin secretion (GSIS), producing glucose intolerance. We showed that testosterone action on AR in β-cells amplifies the insulinotropic action of GLP-1 on its receptor via a cAMP-dependent protein kinase-A pathway. METHODS To investigate AR-dependent gene networks in β-cells, we performed a high throughput whole transcriptome sequencing (RNA-Seq) in islets from male βARKO-/y and control mice. RESULTS We identified 214 differentially expressed genes (DEGs) (158 up- and 56 down-regulated) with a false discovery rate (FDR) < 0.05 and a fold change (FC) > 2. Our analysis of individual transcripts revealed alterations in β-cell genes involved in cellular inflammation/stress and insulin secretion. Based on 312 DEGs with an FDR < 0.05, the pathway analysis revealed 23 significantly enriched pathways, including cytokine-cytokine receptor interaction, Jak-STAT signaling, insulin signaling, MAPK signaling, type 2 diabetes (T2D) and pancreatic secretion. The gene ontology analysis confirmed the results of the individual DEGs and the pathway analysis in showing enriched biological processes encompassing inflammation, ion transport, exocytosis and insulin secretion. CONCLUSIONS AR-deficient islets exhibit altered expression of genes involved in inflammation and insulin secretion demonstrating the importance of androgen action in β-cell health in the male with implications for T2D development in men.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Crosses, Genetic
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Gene Expression Profiling
- Gene Expression Regulation
- Gene Ontology
- Genetic Markers
- High-Throughput Nucleotide Sequencing
- Insulin/metabolism
- Insulin Secretion
- Insulin-Secreting Cells/immunology
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/pathology
- Islets of Langerhans/immunology
- Islets of Langerhans/metabolism
- Islets of Langerhans/pathology
- Male
- Mice
- Mice, Knockout
- Mice, Transgenic
- Models, Biological
- Organ Specificity
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Sequence Analysis, RNA
- Transcriptome
Collapse
Affiliation(s)
- Weiwei Xu
- Diabetes Discovery Research and Gender Medicine Laboratory, Department of Medicine, Section of Endocrinology and Metabolism, Tulane University School of Medicine, New Orleans
| | - Tianhua Niu
- Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA
| | - Beibei Xu
- Diabetes Discovery Research and Gender Medicine Laboratory, Department of Medicine, Section of Endocrinology and Metabolism, Tulane University School of Medicine, New Orleans
| | - Guadalupe Navarro
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Matthew J Schipma
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Franck Mauvais-Jarvis
- Diabetes Discovery Research and Gender Medicine Laboratory, Department of Medicine, Section of Endocrinology and Metabolism, Tulane University School of Medicine, New Orleans.
| |
Collapse
|
78
|
Siglec-7 restores β-cell function and survival and reduces inflammation in pancreatic islets from patients with diabetes. Sci Rep 2017; 7:45319. [PMID: 28378743 PMCID: PMC5381285 DOI: 10.1038/srep45319] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 02/22/2017] [Indexed: 01/16/2023] Open
Abstract
Chronic inflammation plays a key role in both type 1 and type 2 diabetes. Cytokine and chemokine production within the islets in a diabetic milieu results in β-cell failure and diabetes progression. Identification of targets, which both prevent macrophage activation and infiltration into islets and restore β-cell functionality is essential for effective diabetes therapy. We report that certain Sialic-acid-binding immunoglobulin-like-lectins (siglecs) are expressed in human pancreatic islets in a cell-type specific manner. Siglec-7 was expressed on β-cells and down-regulated in type 1 and type 2 diabetes and in infiltrating activated immune cells. Over-expression of Siglec-7 in diabetic islets reduced cytokines, prevented β-cell dysfunction and apoptosis and reduced recruiting of migrating monocytes. Our data suggest that restoration of human Siglec-7 expression may be a novel therapeutic strategy targeted to both inhibition of immune activation and preservation of β-cell function and survival.
Collapse
|
79
|
Wang HJ, Zhou Y, Liu RM, Qin YS, Cen YH, Hu LY, Wang SM, Hu ZJ. IP-10/CXCR3 Axis Promotes the Proliferation of Vascular Smooth Muscle Cells through ERK1/2/CREB Signaling Pathway. Cell Biochem Biophys 2017; 75:139-147. [PMID: 28111710 DOI: 10.1007/s12013-017-0782-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 01/11/2017] [Indexed: 10/20/2022]
Abstract
Excessive proliferation of vascular smooth muscle cells is one of the main pathological processes leading to atherosclerosis and intimal hyperplasia after vascular interventional therapy. Our previous study has shown that interferon-γ inducible protein-10 contributes to the proliferation of vascular smooth muscle cell. However, the underlying mechanisms remain unclear. Extracellular signal-regulated kinase 1/2, serine/threonine kinase Akt, and cAMP response element binding protein are signaling pathways, which are considered to play important roles in the processes of vascular smooth muscle cell proliferation. Moreover, chemokine receptor 3 and Toll-like receptor 4 are potential receptors of inducible protein-10 in this process. In the present study, IP-10 was found to directly induce vascular smooth muscle cell proliferation, and exposure to inducible protein-10 activated extracellular signal-regulated kinase 1/2, serine/threonine kinase, and cAMP response element binding protein signaling. Inhibitor of extracellular signal-regulated kinase 1/2, rather than inhibitor of serine/threonine kinase, inhibited the phosphorylation of cAMP response element binding protein and reduced inducible protein-10-stimulated vascular smooth muscle cell proliferation. Knockdown of cAMP response element binding protein by siRNA inhibited inducible protein-10-induced vascular smooth muscle cell proliferation. Moreover, anti-CXCR3 IgG, instead of anti-Toll-like receptor 4 IgG, reduced inducible protein-10-induced vascular smooth muscle cell proliferation and inducible protein-10-stimulated extracellular signal-regulated kinase 1/2 and cAMP response element binding protein activation. Together, these results indicate that inducible protein-10 promotes vascular smooth muscle cell proliferation via chemokine receptor 3 and activation of extracellular signal-regulated kinase 1/2 inducible protein-10-induced vascular smooth muscle cell proliferation. These data provide important targets for future studies to modulate atherosclerosis and restenosis after vascular interventional therapy.
Collapse
Affiliation(s)
- Hui-Jin Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.,Department of General Surgery, Huadu District People's Hospital, Southern Medical University, Guanghzou, 510800, China
| | - Yu Zhou
- Department of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Rui-Ming Liu
- Laboratory of Department of Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yuan-Sen Qin
- Department of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Ying-Huan Cen
- Department of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Ling-Yu Hu
- Department of Breast Surgery, Guangzhou Women and Children's Medical Center, Guangzhou, 510080, China
| | - Shen-Ming Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Zuo-Jun Hu
- Department of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
80
|
Abstract
INTRODUCTION By virtue of its specificity for chemokines induced in Th1-associated pathologies, CXCR3 has attracted considerable attention as a target for therapeutic intervention. Several pharmacologically distinct small molecules with in vitro and in vivo potency have been described in the literature, although to date, none have shown efficacy in clinical trials. Areas covered: In this article, the author outlines the rationale for targeting CXCR3 and discusses the potential pitfalls in targeting receptors in poorly understood areas of chemokine biology. Furthermore, they cover emerging therapeutic areas outside of the 'traditional' Th1 arena in which CXCR3 antagonists may ultimately bear fruit. Finally, they discuss the design of recently discovered small molecules targeting CXCR3. Expert opinion: CXCR3 and its ligands appear to play roles in a multitude of diverse diseases in humans. In vitro studies suggest that CXCR3 is inherently 'druggable' and that potent, efficacious small molecules targeting CXCR3 antagonists will find a clinical niche. However, the well-trodden path to failure of small molecule chemokine receptor antagonists in clinical trials suggests that a cautious approach should be undertaken. Ideally, unequivocal evidence elucidating the precise role of CXCR3 should be obtained before targeting the receptor in a particular disease cohort.
Collapse
Affiliation(s)
- James E Pease
- a Inflammation, Repair & Development Section, National Heart & Lung Institute, Faculty of Medicine , Imperial College London , London , UK
| |
Collapse
|
81
|
Rehman K, Akash MSH. Mechanisms of inflammatory responses and development of insulin resistance: how are they interlinked? J Biomed Sci 2016; 23:87. [PMID: 27912756 PMCID: PMC5135788 DOI: 10.1186/s12929-016-0303-y] [Citation(s) in RCA: 298] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 11/24/2016] [Indexed: 02/06/2023] Open
Abstract
Background Insulin resistance (IR) is one of the major hallmark for pathogenesis and etiology of type 2 diabetes mellitus (T2DM). IR is directly interlinked with various inflammatory responses which play crucial role in the development of IR. Inflammatory responses play a crucial role in the pathogenesis and development of IR which is one of the main causative factor for the etiology of T2DM. Methods A comprehensive online English literature was searched using various electronic search databases. Different search terms for pathogenesis of IR, role of various inflammatory responses were used and an advanced search was conducted by combining all the search fields in abstracts, keywords, and titles. Results We summarized the data from the searched articles and found that inflammatory responses activate the production of various pro-inflammatory mediators notably cytokines, chemokines and adipocytokines through the involvement of various transcriptional mediated molecular pathways, oxidative and metabolic stress. Overnutrition is one of the major causative factor that contributes to induce the state of low-grade inflammation due to which accumulation of elevated levels of glucose and/or lipids in blood stream occur that leads to the activation of various transcriptional mediated molecular and metabolic pathways. This results in the induction of various pro-inflammatory mediators that are decisively involved to provoke the pathogenesis of tissue-specific IR by interfering with insulin signaling pathways. Once IR is developed, it increases oxidative stress in β-cells of pancreatic islets and peripheral tissues which impairs insulin secretion, and insulin sensitivity in β-cells of pancreatic islets and peripheral tissues, respectively. Moreover, we also summarized the data regarding various treatment strategies of inflammatory responses-induced IR. Conclusions In this article, we have briefly described that how pro-inflammatory mediators, oxidative stress, transcriptional mediated molecular and metabolic pathways are involved in the pathogenesis of tissues-specific IR. Moreover, based on recent investigations, we have also described that to counterfeit these inflammatory responses is one of the best treatment strategy to prevent the pathogenesis of IR through ameliorating the incidences of inflammatory responses.
Collapse
Affiliation(s)
- Kanwal Rehman
- Institute of Pharmacy, Physiology and Pharmacology, University of Agriculture, Faisalabad, Pakistan
| | | |
Collapse
|
82
|
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]
|
83
|
Yuan T, Rafizadeh S, Azizi Z, Lupse B, Gorrepati KDD, Awal S, Oberholzer J, Maedler K, Ardestani A. Proproliferative and antiapoptotic action of exogenously introduced YAP in pancreatic β cells. JCI Insight 2016; 1:e86326. [PMID: 27812538 DOI: 10.1172/jci.insight.86326] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Loss of functional pancreatic β cells is a hallmark of both type 1 and 2 diabetes. Identifying the pathways that promote β cell proliferation and/or block β cell apoptosis is a potential strategy for diabetes therapy. The transcriptional coactivator Yes-associated protein (YAP), a major downstream effector of the Hippo signaling pathway, is a key regulator of organ size and tissue homeostasis by modulating cell proliferation and apoptosis. YAP is not expressed in mature primary human and mouse β cells. We aimed to identify whether reexpression of a constitutively active form of YAP promotes β cell proliferation/survival. Overexpression of YAP remarkably induced β cell proliferation in isolated human islets, while β cell function and functional identity genes were fully preserved. The transcription factor forkhead box M1 (FOXM1) was upregulated upon YAP overexpression and necessary for YAP-dependent β cell proliferation. YAP overexpression protected β cells from apoptosis triggered by multiple diabetic conditions. The small redox proteins thioredoxin-1 and thioredoxin-2 (Trx1/2) were upregulated by YAP; disruption of the Trx system revealed that Trx1/2 was required for the antiapoptotic action of YAP in insulin-producing β cells. Our data show the robust proproliferative and antiapoptotic function of YAP in pancreatic β cells. YAP reconstitution may represent a disease-modifying approach to restore a functional β cell mass in diabetes.
Collapse
Affiliation(s)
- Ting Yuan
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Sahar Rafizadeh
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Zahra Azizi
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Blaz Lupse
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | | | - Sushil Awal
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Jose Oberholzer
- Division of Transplantation, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Kathrin Maedler
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany.,German Center for Diabetes Research (DZD) project partner, University of Bremen, Bremen, Germany
| | - Amin Ardestani
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| |
Collapse
|
84
|
Lundberg M, Krogvold L, Kuric E, Dahl-Jørgensen K, Skog O. Expression of Interferon-Stimulated Genes in Insulitic Pancreatic Islets of Patients Recently Diagnosed With Type 1 Diabetes. Diabetes 2016; 65:3104-10. [PMID: 27422384 DOI: 10.2337/db16-0616] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/08/2016] [Indexed: 11/13/2022]
Abstract
A primary insult to the pancreatic islets of Langerhans, leading to the activation of innate immunity, has been suggested as an important step in the inflammatory process in type 1 diabetes (T1D). The aim of this study was to examine whether interferon (IFN)-stimulated genes (ISGs) are overexpressed in human T1D islets affected with insulitis. By using laser capture microdissection and a quantitative PCR array, 23 of 84 examined ISGs were found to be overexpressed by at least fivefold in insulitic islets from living patients with recent-onset T1D, participating in the Diabetes Virus Detection (DiViD) study, compared with islets from organ donors without diabetes. Most of the overexpressed ISGs, including GBP1, TLR3, OAS1, EIF2AK2, HLA-E, IFI6, and STAT1, showed higher expression in the islet core compared with the peri-islet area containing the surrounding immune cells. In contrast, the T-cell attractant chemokine CXCL10 showed an almost 10-fold higher expression in the peri-islet area than in the islet, possibly partly explaining the localization of T cells mainly to this region. In conclusion, insulitic islets from recent-onset T1D subjects show overexpression of ISGs, with an expression pattern similar to that seen in islets infected with virus or exposed to IFN-γ/interleukin-1β or IFN-α.
Collapse
Affiliation(s)
- Marcus Lundberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Lars Krogvold
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Enida Kuric
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Knut Dahl-Jørgensen
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Oskar Skog
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
85
|
C-X-C motif chemokine 10 in non-alcoholic steatohepatitis: role as a pro-inflammatory factor and clinical implication. Expert Rev Mol Med 2016; 18:e16. [PMID: 27669973 DOI: 10.1017/erm.2016.16] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease. Non-alcoholic steatohepatitis (NASH) is a more severe form of NAFLD and causes subsequent pathological changes including cirrhosis and hepatocellular carcinoma. Inflammation is the key pathological change in NASH and involves a series of cytokines and chemokines. The C-X-C motif chemokine 10 (CXCL10), which is known as a pro-inflammation chemokine, was recently proven to play a pivotal role in the pathogenesis of NASH. Hepatic CXCL10 is mainly secreted by hepatocytes and liver sinusoidal endothelium. By binding to its specific receptor CXCR3, CXCL10 recruits activated CXCR3+ T lymphocytes and macrophages to parenchyma and promotes inflammation, apoptosis and fibrosis. The circulating CXCL10 level correlates with the severity of lobular inflammation and is an independent risk factor for NASH patients. Thus, CXCL10 may be both a potential prognostic tool and a therapeutic target for the treatment of patients with NASH. The aim of this review is to highlight the growing advances in basic knowledge and clinical interest of CXCL10 in NASH to propagate new insights into novel pharmacotherapeutic avenues.
Collapse
|
86
|
Shah P, Lueschen N, Ardestani A, Oberholzer J, Olerud J, Carlsson PO, Maedler K. Angiopoetin-2 Signals Do Not Mediate the Hypervascularization of Islets in Type 2 Diabetes. PLoS One 2016; 11:e0161834. [PMID: 27617438 PMCID: PMC5019443 DOI: 10.1371/journal.pone.0161834] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Accepted: 08/12/2016] [Indexed: 12/12/2022] Open
Abstract
AIMS Changes in the islet vasculature have been implicated in the regulation of β-cell survival and function during the progression to type 2 diabetes (T2D). Failure of the β-cell to compensate for the increased insulin demand in obesity eventually leads to diabetes; as a result of the complex interplay of genetic and environmental factors (e.g. ongoing inflammation within the islets) and impaired vascular function. The Angiopoietin/Tie (Ang/Tie) angiogenic system maintains vasculature and is closely related to organ inflammation and angiogenesis. In this study we aimed to identify whether the vessel area within the islets changes in diabetes and whether such changes would be triggered by the Tie-antagonist Ang-2. METHODS Immunohistochemical and qPCR analyses to follow islet vascularization and Ang/Tie levels were performed in human pancreatic autopsies and isolated human and mouse islets. The effect of Ang-2 was assessed in β-cell-specific Ang-2 overexpressing mice during high fat diet (HFD) feeding. RESULTS Islet vessel area was increased in autopsy pancreases from patients with T2D. The vessel markers Tie-1, Tie-2 and CD31 were upregulated in mouse islets upon HFD feeding from 8 to 24 weeks. Ang-2 was transiently upregulated in mouse islets at 8 weeks of HFD and under glucolipotoxic conditions (22.2 mM glucose/ 0.5 mM palmitate) in vitro in human and mouse islets, in contrast to its downregulation by cytokines (IL-1β, IFN-ɣ and TNF-α). Ang-1 on the other hand was oppositely regulated, with a significant loss under glucolipotoxic condition, a trend to reduce in islets from patients with T2D and an upregulation by cytokines. Modulation of such changes in Ang-2 by its overexpression or the inhibition of its receptor Tie-2 impaired β-cell function at basal conditions but protected islets from cytokine induced apoptosis. In vivo, β-cell-specific Ang-2 overexpression in mice induced hypervascularization under normal diet but contrastingly led to hypovascularized islets in response to HFD together with increased apoptosis and reduced β-cell mass. CONCLUSIONS Islet hypervascularization occurs in T2D. A balanced expression of the Ang1/Ang2 system is important for islet physiology. Ang-2 prevents β-cell mass and islet vascular adaptation in response to HFD feeding with no major influence on glucose homeostasis.
Collapse
Affiliation(s)
- Payal Shah
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany
| | - Navina Lueschen
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany
| | - Amin Ardestani
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany
| | - Jose Oberholzer
- Division of Transplantation, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Johan Olerud
- Department of Immunology, Genetics and pathology, Uppsala University, Uppsala, Sweden
| | - Per-Ola Carlsson
- Department of Medical cell biology and Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Kathrin Maedler
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany,German Center for Diabetes Research (DZD) project partner, University of Bremen, Bremen, Germany,* E-mail:
| |
Collapse
|
87
|
CXCL10-Mediates Macrophage, but not Other Innate Immune Cells-Associated Inflammation in Murine Nonalcoholic Steatohepatitis. Sci Rep 2016; 6:28786. [PMID: 27349927 PMCID: PMC4923862 DOI: 10.1038/srep28786] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/10/2016] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is an inflammatory lipotoxic disorder, but how inflammatory cells are recruited and activated within the liver is still unclear. We previously reported that lipotoxic hepatocytes release CXCL10-enriched extracellular vesicles, which are potently chemotactic for cells of the innate immune system. In the present study, we sought to determine the innate immune cell involved in the inflammatory response in murine NASH and the extent to which inhibition of the chemotactic ligand CXCL10 and its cognate receptor CXCR3 could attenuate liver inflammation, injury and fibrosis. C57BL/6J CXCL10−/−, CXCR3−/− and wild type (WT) mice were fed chow or high saturated fat, fructose, and cholesterol (FFC) diet. FFC-fed CXCL10−/− and WT mice displayed similar weight gain, metabolic profile, insulin resistance, and hepatic steatosis. In contrast, compared to the WT mice, FFC-fed CXCL10−/− mice had significantly attenuated liver inflammation, injury and fibrosis. Genetic deletion of CXCL10 reduced FFC-induced proinflammatory hepatic macrophage infiltration, while natural killer cells, natural killer T cells, neutrophils and dendritic cells hepatic infiltration were not significantly affected. Our results suggest that CXCL10−/− mice are protected against diet-induced NASH, in an obesity-independent manner. Macrophage-associated inflammation appears to be the key player in the CXCL10-mediated sterile inflammatory response in murine NASH.
Collapse
|
88
|
Lekva T, Norwitz ER, Aukrust P, Ueland T. Impact of Systemic Inflammation on the Progression of Gestational Diabetes Mellitus. Curr Diab Rep 2016; 16:26. [PMID: 26879309 DOI: 10.1007/s11892-016-0715-9] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
With increasing rates of obesity and new diagnostic criteria for gestational diabetes mellitus (GDM), the overall prevalence of GDM is increasing worldwide. Women with GDM have an increased risk of maternal and fetal complications during pregnancy as well as long-term risks including higher prevalence of type 2 diabetes mellitus and cardiovascular disease. In recent years, the role of immune activation and inflammation in the pathogenesis of GDM has gained increasing attention. This monograph explores the current state of the literature as regards the expression of markers of inflammation in the maternal circulation, placenta, and adipose tissue of women with GDM.
Collapse
Affiliation(s)
- Tove Lekva
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Sognsvannsveien 20, 0027, Oslo, Norway.
- Mother Infant Research Institute, Tufts Medical Center, 800 Washington Street, Boston, MA, 02116, USA.
| | - Errol R Norwitz
- Mother Infant Research Institute, Tufts Medical Center, 800 Washington Street, Boston, MA, 02116, USA.
- Department of Obstetrics & Gynecology, Tufts Medical Center and Tufts University School of Medicine, Boston, MA, USA.
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Sognsvannsveien 20, 0027, Oslo, Norway.
- Faculty of Medicine, University of Oslo, Oslo, Norway.
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway.
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Sognsvannsveien 20, 0027, Oslo, Norway.
- Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
89
|
Prause M, Berchtold LA, Urizar AI, Hyldgaard Trauelsen M, Billestrup N, Mandrup-Poulsen T, Størling J. TRAF2 mediates JNK and STAT3 activation in response to IL-1β and IFNγ and facilitates apoptotic death of insulin-producing β-cells. Mol Cell Endocrinol 2016; 420:24-36. [PMID: 26610752 DOI: 10.1016/j.mce.2015.11.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 11/16/2015] [Accepted: 11/16/2015] [Indexed: 12/01/2022]
Abstract
Interleukin-1β (IL-1β) and interferon-γ (IFNγ) contribute to type 1 diabetes (T1D) by inducing β-cell death. Tumor necrosis factor (TNF) receptor-associated factor (TRAF) proteins are adaptors that transduce signaling from a variety of membrane receptors including cytokine receptors. We show here that IL-1β and IFNγ upregulate the expression of TRAF2 in insulin-producing INS-1E cells and isolated rat pancreatic islets. siRNA-mediated knockdown (KD) of TRAF2 in INS-1E cells reduced IL-1β-induced phosphorylation of JNK1/2, but not of p38 or ERK1/2 mitogen-activated protein kinases. TRAF2 KD did not modulate NFκB activation by cytokines, but reduced cytokine-induced inducible nitric oxide synthase (iNOS) promotor activity and expression. We further observed that IFNγ-stimulated phosphorylation of STAT3 required TRAF2. KD of TRAF2 or STAT3 reduced cytokine-induced caspase 3/7 activation, but, intriguingly, potentiated cytokine-mediated loss of plasma membrane integrity and augmented the number of propidium iodide-positive cells. Finally, we found that TRAF2 KD increased cytokine-induced production of reactive oxygen species (ROS). In summary, our data suggest that TRAF2 is an important mediator of IL-1β and IFNγ signaling in pancreatic β-cells.
Collapse
Affiliation(s)
- Michala Prause
- Immunoendocrinology Laboratory, Endocrinology Research Section, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark; Section of Cellular and Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lukas Adrian Berchtold
- Section of Cellular and Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Adriana Ibarra Urizar
- Section of Cellular and Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette Hyldgaard Trauelsen
- Beta-Cell Biology Group, Copenhagen Diabetes Research Center, Department of Paediatrics E, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Nils Billestrup
- Section of Cellular and Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Mandrup-Poulsen
- Immunoendocrinology Laboratory, Endocrinology Research Section, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Joachim Størling
- Beta-Cell Biology Group, Copenhagen Diabetes Research Center, Department of Paediatrics E, Copenhagen University Hospital Herlev, Herlev, Denmark.
| |
Collapse
|
90
|
Kaur S, Mirza AH, Brorsson CA, Fløyel T, Størling J, Mortensen HB, Pociot F. The genetic and regulatory architecture of ERBB3-type 1 diabetes susceptibility locus. Mol Cell Endocrinol 2016; 419:83-91. [PMID: 26450151 DOI: 10.1016/j.mce.2015.10.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/29/2015] [Accepted: 10/01/2015] [Indexed: 12/11/2022]
Abstract
The study aimed to explore the role of ERBB3 in type 1 diabetes (T1D). We examined whether genetic variation of ERBB3 (rs2292239) affects residual β-cell function in T1D cases. Furthermore, we examined the expression of ERBB3 in human islets, the effect of ERBB3 knockdown on apoptosis in insulin-producing INS-1E cells and the genetic and regulatory architecture of the ERBB3 locus to provide insights to how rs2292239 may confer disease susceptibility. rs2292239 strongly correlated with residual β-cell function and metabolic control in children with T1D. ERBB3 locus associated lncRNA (NONHSAG011351) was found to be expressed in human islets. ERBB3 was expressed and down-regulated by pro-inflammatory cytokines in human islets and INS-1E cells; knockdown of ERBB3 in INS-1E cells decreased basal and cytokine-induced apoptosis. Our data suggests an important functional role of ERBB3 and its potential regulators in the β-cells and may constitute novel targets to prevent β-cell destruction in T1D.
Collapse
Affiliation(s)
- Simranjeet Kaur
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Pediatrics, Herlev University Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Aashiq H Mirza
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Pediatrics, Herlev University Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Center for Non-coding RNA in Technology and Health, University of Copenhagen, Denmark
| | - Caroline A Brorsson
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Pediatrics, Herlev University Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark
| | - Tina Fløyel
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Pediatrics, Herlev University Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark
| | - Joachim Størling
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Pediatrics, Herlev University Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark
| | - Henrik B Mortensen
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Pediatrics, Herlev University Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Flemming Pociot
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Pediatrics, Herlev University Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Center for Non-coding RNA in Technology and Health, University of Copenhagen, Denmark.
| |
Collapse
|
91
|
Affiliation(s)
- Dirk Homann
- Diabetes, Obesity and Metabolism Institute and Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
92
|
Van Raemdonck K, Van den Steen PE, Liekens S, Van Damme J, Struyf S. CXCR3 ligands in disease and therapy. Cytokine Growth Factor Rev 2015; 26:311-27. [DOI: 10.1016/j.cytogfr.2014.11.009] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 11/05/2014] [Indexed: 12/19/2022]
|
93
|
Meyer A, Stolz K, Dreher W, Bergemann J, Holebasavanahalli Thimmashetty V, Lueschen N, Azizi Z, Khobragade V, Maedler K, Kuestermann E. Manganese-mediated MRI signals correlate with functional β-cell mass during diabetes progression. Diabetes 2015; 64:2138-47. [PMID: 25804940 DOI: 10.2337/db14-0864] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 01/13/2015] [Indexed: 11/13/2022]
Abstract
Diabetes diagnostic therapy and research would strongly benefit from noninvasive accurate imaging of the functional β-cells in the pancreas. Here, we developed an analysis of functional β-cell mass (BCM) by measuring manganese (Mn(2+)) uptake kinetics into glucose-stimulated β-cells by T1-weighted in vivo Mn(2+)-mediated MRI (MnMRI) in C57Bl/6J mice. Weekly MRI analysis during the diabetes progression in mice fed a high-fat/high-sucrose diet (HFD) showed increased Mn(2+)-signals in the pancreas of the HFD-fed mice during the compensation phase, when glucose tolerance and glucose-stimulated insulin secretion (GSIS) were improved and BCM was increased compared with normal diet-fed mice. The increased signal was only transient; from the 4th week on, MRI signals decreased significantly in the HFD group, and the reduced MRI signal in HFD mice persisted over the whole 12-week experimental period, which again correlated with both impaired glucose tolerance and GSIS, although BCM remained unchanged. Rapid and significantly decreased MRI signals were confirmed in diabetic mice after streptozotocin (STZ) injection. No long-term effects of Mn(2+) on glucose tolerance were observed. Our optimized MnMRI protocol fulfills the requirements of noninvasive MRI analysis and detects already small changes in the functional BCM.
Collapse
Affiliation(s)
- Anke Meyer
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Katharina Stolz
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | | | - Jennifer Bergemann
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | | | - Navina Lueschen
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Zahra Azizi
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Vrushali Khobragade
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Kathrin Maedler
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | | |
Collapse
|
94
|
Transcriptional regulation of chemokine genes: a link to pancreatic islet inflammation? Biomolecules 2015; 5:1020-34. [PMID: 26018641 PMCID: PMC4496708 DOI: 10.3390/biom5021020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 05/12/2015] [Indexed: 12/18/2022] Open
Abstract
Enhanced expression of chemotactic cytokines (aka chemokines) within pancreatic islets likely contributes to islet inflammation by regulating the recruitment and activation of various leukocyte populations, including macrophages, neutrophils, and T-lymphocytes. Because of the powerful actions of these chemokines, precise transcriptional control is required. In this review, we highlight what is known about the signals and mechanisms that govern the transcription of genes encoding specific chemokine proteins in pancreatic islet β-cells, which include contributions from the NF-κB and STAT1 pathways. We further discuss increased chemokine expression in pancreatic islets during autoimmune-mediated and obesity-related development of diabetes.
Collapse
|
95
|
Chang CC, Wu CL, Su WW, Shih KL, Tarng DC, Chou CT, Chen TY, Kor CT, Wu HM. Interferon gamma-induced protein 10 is associated with insulin resistance and incident diabetes in patients with nonalcoholic fatty liver disease. Sci Rep 2015; 5:10096. [PMID: 25961500 PMCID: PMC4426720 DOI: 10.1038/srep10096] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 03/30/2015] [Indexed: 12/11/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is an important risk factor for the development of type 2 diabetes mellitus. Interferon gamma-induced protein 10 (IP-10), a proinflammatory chemokine, plays a crucial role in inflammatory diseases. This cross-sectional pilot study investigated whether circulating IP-10 is associated with the progression of liver disease, and prediabetes in patients with NAFLD. A total of 90 patients with NAFLD alone (n = 48) or NAFLD with incident diabetes (n = 42) and 43 controls participated in this study. Fasting plasma was used to assess metabolic parameters, inflammatory factors, endotoxin levels, and malondialdehyde (MDA) concentrations. Insulin resistance was estimated using homeostatic model assessment (HOMA-IR). IP-10 levels were significantly higher in patients with NAFLD alone (median (interquartile range): 369.44 (309.30-418.97) pg/mL) and in those with incident diabetes (418.99 (330.73-526.04) pg/mL) than in controls (293.37 (214.10-331.57) pg/mL) (P < 0.001). IP-10 levels were positively correlated with levels of alanine aminotransferase, hs-CRP, MDA, MCP-1, and TNF-α as well as HOMA-IR values. Ordinal logistic regression analysis revealed IP-10 was an independent risk factor associated with progressive liver injury, insulin resistance and incident diabetes. Circulating IP-10 may be a non-invasive biomarker for disease progression and subsequent diabetes development of NAFLD.
Collapse
Affiliation(s)
- Chia-Chu Chang
- 1] Division of Nephrology, Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan [2] School of Medicine, Chung-Shan Medical University, Taichung, Taiwan
| | - Chia-Lin Wu
- 1] Division of Nephrology, Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan [2] School of Medicine, Chung-Shan Medical University, Taichung, Taiwan [3] Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Wei-Wen Su
- Department of Gastroenterology, Changhua Christian Hospital, Changhua, Taiwan
| | - Kai-Lun Shih
- Department of Gastroenterology, Changhua Christian Hospital, Changhua, Taiwan
| | - Der-Cherng Tarng
- 1] Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan [2] Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan [3] Department and Institute of Physiology, National Yang-Ming University, Taipei, Taiwan
| | - Chen-Te Chou
- Department of Medical Imaging, Changhua Christian Hospital, Changhua, Taiwan
| | - Ting-Yu Chen
- Inflammation Research &Drug Development Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Chew-Teng Kor
- Internal Medicine Research Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Hung-Ming Wu
- 1] Inflammation Research &Drug Development Center, Changhua Christian Hospital, Changhua, Taiwan [2] Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan [3] Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan
| |
Collapse
|
96
|
Bonner C, Kerr-Conte J, Gmyr V, Queniat G, Moerman E, Thévenet J, Beaucamps C, Delalleau N, Popescu I, Malaisse WJ, Sener A, Deprez B, Abderrahmani A, Staels B, Pattou F. Inhibition of the glucose transporter SGLT2 with dapagliflozin in pancreatic alpha cells triggers glucagon secretion. Nat Med 2015; 21:512-7. [PMID: 25894829 DOI: 10.1038/nm.3828] [Citation(s) in RCA: 471] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 02/19/2015] [Indexed: 12/11/2022]
Abstract
Type 2 diabetes (T2D) is characterized by chronic hyperglycemia resulting from a deficiency in insulin signaling, because of insulin resistance and/or defects in insulin secretion; it is also associated with increases in glucagon and endogenous glucose production (EGP). Gliflozins, including dapagliflozin, are a new class of approved oral antidiabetic agents that specifically inhibit sodium-glucose co-transporter 2 (SGLT2) function in the kidney, thus preventing renal glucose reabsorption and increasing glycosuria in diabetic individuals while reducing hyperglycemia. However, gliflozin treatment in subjects with T2D increases both plasma glucagon and EGP by unknown mechanisms. In spite of the rise in EGP, T2D patients treated with gliflozin have lower blood glucose levels than those receiving placebo, possibly because of increased glycosuria; however, the resulting increase in plasma glucagon levels represents a possible concerning side effect, especially in a patient population already affected by hyperglucagonemia. Here we demonstrate that SGLT2 is expressed in glucagon-secreting alpha cells of the pancreatic islets. We further found that expression of SLC5A2 (which encodes SGLT2) was lower and glucagon (GCG) gene expression was higher in islets from T2D individuals and in normal islets exposed to chronic hyperglycemia than in islets from non-diabetics. Moreover, hepatocyte nuclear factor 4-α (HNF4A) is specifically expressed in human alpha cells, in which it controls SLC5A2 expression, and its expression is downregulated by hyperglycemia. In addition, inhibition of either SLC5A2 via siRNA-induced gene silencing or SGLT2 via dapagliflozin treatment in human islets triggered glucagon secretion through KATP channel activation. Finally, we found that dapagliflozin treatment further promotes glucagon secretion and hepatic gluconeogenesis in healthy mice, thereby limiting the decrease of plasma glucose induced by fasting. Collectively, these results identify a heretofore unknown role of SGLT2 and designate dapagliflozin an alpha cell secretagogue.
Collapse
Affiliation(s)
- Caroline Bonner
- 1] European Genomic Institute for Diabetes, Lille, France. [2] INSERM UMR 1190, Lille, France. [3] Centre Hospitalier Régional Universitaire, Lille, France
| | - Julie Kerr-Conte
- 1] European Genomic Institute for Diabetes, Lille, France. [2] INSERM UMR 1190, Lille, France. [3] Centre Hospitalier Régional Universitaire, Lille, France. [4] Université de Lille, Lille, France
| | - Valéry Gmyr
- 1] European Genomic Institute for Diabetes, Lille, France. [2] INSERM UMR 1190, Lille, France. [3] Université de Lille, Lille, France
| | - Gurvan Queniat
- 1] European Genomic Institute for Diabetes, Lille, France. [2] INSERM UMR 1190, Lille, France. [3] Université de Lille, Lille, France
| | - Ericka Moerman
- 1] European Genomic Institute for Diabetes, Lille, France. [2] INSERM UMR 1190, Lille, France. [3] Université de Lille, Lille, France
| | - Julien Thévenet
- 1] European Genomic Institute for Diabetes, Lille, France. [2] INSERM UMR 1190, Lille, France. [3] Université de Lille, Lille, France
| | - Cédric Beaucamps
- 1] European Genomic Institute for Diabetes, Lille, France. [2] INSERM UMR 1190, Lille, France. [3] Centre Hospitalier Régional Universitaire, Lille, France
| | - Nathalie Delalleau
- 1] European Genomic Institute for Diabetes, Lille, France. [2] INSERM UMR 1190, Lille, France. [3] Université de Lille, Lille, France
| | - Iuliana Popescu
- Laboratory of Experimental Hormonology, Medical School, Université Libre de Bruxelles, Brussels, Belgium
| | - Willy J Malaisse
- Laboratory of Experimental Hormonology, Medical School, Université Libre de Bruxelles, Brussels, Belgium
| | - Abdullah Sener
- Laboratory of Experimental Hormonology, Medical School, Université Libre de Bruxelles, Brussels, Belgium
| | - Benoit Deprez
- 1] Université de Lille, Lille, France. [2] INSERM UMR 1177, Lille, France. [3] Institut Pasteur de Lille, Lille, France
| | - Amar Abderrahmani
- 1] European Genomic Institute for Diabetes, Lille, France. [2] Université de Lille, Lille, France. [3] CNRS UMR 8199, Lille, France
| | - Bart Staels
- 1] European Genomic Institute for Diabetes, Lille, France. [2] Université de Lille, Lille, France. [3] Institut Pasteur de Lille, Lille, France. [4] INSERM UMR 1011, Lille, France
| | - François Pattou
- 1] European Genomic Institute for Diabetes, Lille, France. [2] INSERM UMR 1190, Lille, France. [3] Centre Hospitalier Régional Universitaire, Lille, France. [4] Université de Lille, Lille, France
| |
Collapse
|
97
|
CCL20 is elevated during obesity and differentially regulated by NF-κB subunits in pancreatic β-cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2015; 1849:637-52. [PMID: 25882704 DOI: 10.1016/j.bbagrm.2015.03.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 03/05/2015] [Accepted: 03/30/2015] [Indexed: 12/20/2022]
Abstract
Enhanced leukocytic infiltration into pancreatic islets contributes to inflammation-based diminutions in functional β-cell mass. Insulitis (aka islet inflammation), which can be present in both T1DM and T2DM, is one factor influencing pancreatic β-cell death and dysfunction. IL-1β, an inflammatory mediator in both T1DM and T2DM, acutely (within 1h) induced expression of the CCL20 gene in rat and human islets and clonal β-cell lines. Transcriptional induction of CCL20 required the p65 subunit of NF-κB to replace the p50 subunit at two functional κB sites within the CCL20 proximal gene promoter. The NF-κB p50 subunit prevents CCL20 gene expression during unstimulated conditions and overexpression of p50 reduces CCL20, but enhances cyclooxygenase-2 (COX-2), transcript accumulation after exposure to IL-1β. We also identified differential recruitment of specific co-activator molecules to the CCL20 gene promoter, when compared with the CCL2 and COX2 genes, revealing distinct transcriptional requirements for individual NF-κB responsive genes. Moreover, IL-1β, TNF-α and IFN-γ individually increased the expression of CCR6, the receptor for CCL20, on the surface of human neutrophils. We further found that the chemokine CCL20 is elevated in serum from both genetically obese db/db mice and in C57BL6/J mice fed a high-fat diet. Taken together, these results are consistent with a possible activation of the CCL20-CCR6 axis in diseases with inflammatory components. Thus, interfering with this signaling pathway, either at the level of NF-κB-mediated chemokine production, or downstream receptor activation, could be a potential therapeutic target to offset inflammation-associated tissue dysfunction in obesity and diabetes.
Collapse
|
98
|
Dietary polyherbal supplementation decreases CD3+ cell infiltration into pancreatic islets and prevents hyperglycemia in nonobese diabetic mice. Nutr Res 2015; 35:328-36. [DOI: 10.1016/j.nutres.2014.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 12/17/2014] [Accepted: 12/19/2014] [Indexed: 01/26/2023]
|
99
|
Grzesik WJ, Nadler JL, Machida Y, Nadler JL, Imai Y, Morris MA. Expression pattern of 12-lipoxygenase in human islets with type 1 diabetes and type 2 diabetes. J Clin Endocrinol Metab 2015; 100:E387-95. [PMID: 25532042 PMCID: PMC4333045 DOI: 10.1210/jc.2014-3630] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
CONTEXT Inflammation in the pancreas can cause β-cell stress, leading to diabetes development. Access to human pancreas tissues via the Network for Pancreatic Organ Donors with Diabetes (nPOD) has allowed characterization of pathways leading to this inflammation. OBJECTIVE 12-Lipoxygenase (12-LO) induces inflammation and has been implicated in diabetes development. Our goal was to determine expression of 12-LO in human islets from control, autoantibody-positive, type 1 diabetic, and type 2 diabetic nPOD pancreas donors. DESIGN Pancreas tissues from nPOD donors were examined by immunohistochemistry and immunofluorescence for islet expression of 12-LO in different subsets of islet cells. PARTICIPANTS Donor pancreas samples were obtained from nPOD based on disease status (control, n = 7; autoantibody-positive, n = 8; type 1 diabetic, n = 17; or type 2 diabetic donors, n = 15). MAIN OUTCOME MEASURE Determination of 12-LO expression within human islets served as the main outcome measure, including distinguishing which types of islet cells expressed 12-LO. RESULTS Islets from control participants (nondiabetic) lacked islet expression of 12-LO. Of donors in the other groups, 25% to 37% expressed islet 12-LO with a clear inverse relation between the numbers of β-cells and 12-LO(+) cells within islets of 12-LO(+) cases. 12-LO expression was not seen within macrophages, endothelial cells, α-cells, or β-cells, but only within cells expressing low levels of pancreatic polypeptide (PP) and increased levels of vimentin. CONCLUSIONS 12-LO expression colocalizes within a specific type of islet PP(+) cell under prediabetic and diabetic conditions. The costaining of PP and vimentin suggests that 12-LO participates in the process leading to β-cell dedifferentiation in the islet.
Collapse
|
100
|
Wu L, Chen X, Mei Y, Hong Q, Feng Z, Lv Y, Wen J, Liu X, Cai G, Chen X. CXCL10 expression induced by Mxi1 inactivation induces mesangial cell apoptosis in mouse Habu nephritis. Cell Signal 2015; 27:943-50. [PMID: 25683914 DOI: 10.1016/j.cellsig.2015.01.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 01/13/2015] [Accepted: 01/27/2015] [Indexed: 11/18/2022]
Abstract
MAX interactor 1 (Mxi1) proteins are c-myc antagonists that primarily exert their biological functions by inhibiting Myc-dependent gene transcription. In this study, Mxi1(-/-) mice were used to generate a model of mesangial proliferative glomerulonephritis for the first time. In the present study, we demonstrated that Mxi1(-/-) mice exhibited a more typical and severe pathological phenotype, which was displayed primarily as a noticeable dissolution phenotype with a higher proportion of apoptotic cells and higher chemokine CXCL10 expression during the early days of modeling, compared with wild-type mice. Additionally, we determined that IRF3-mediated TLR4 signaling was likely involved in regulating CXCL10 expression, which might participate in the mesangial dissolution process. We also found increases in CXCL10 expression, caspase 3 activation, and the proportion of apoptotic cells when Mxi1 expression was inhibited in mouse mesangial cells. Furthermore, the proportion of apoptotic cells decreased after inhibiting CXCL10 expression. Therefore, we concluded that the mesangial cell apoptosis observed in this mesangial proliferative glomerulonephritis model was related to CXCL10 expression induced by Mxi1 inactivation. This finding provides a new theoretical basis for the mechanism of mesangial proliferative glomerulonephritis progression and reveals potential intervention targets for the early treatment of this disease.
Collapse
Affiliation(s)
- Lingling Wu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, China; Medical College, NanKai University, Tianjin, China
| | - Xiaoniao Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, China; Medical College, NanKai University, Tianjin, China
| | - Yan Mei
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, China
| | - Quan Hong
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, China
| | - Zhe Feng
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, China
| | - Yang Lv
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, China
| | - Jun Wen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, China
| | - Xiaoluan Liu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, China
| | - Guangyan Cai
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, China.
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, China.
| |
Collapse
|