1
|
Casas R, Tompa A, Åkesson K, Teixeira PF, Lindqvist A, Ludvigsson J. Redosing with Intralymphatic GAD-Alum in the Treatment of Type 1 Diabetes: The DIAGNODE-B Pilot Trial. Int J Mol Sci 2025; 26:374. [PMID: 39796229 PMCID: PMC11720063 DOI: 10.3390/ijms26010374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Immunotherapies aimed at preserving residual beta cell function in type 1 diabetes have been successful, although the effect has been limited, or raised safety concerns. Transient effects often observed may necessitate redosing to prolong the effect, although this is not always feasible or safe. Treatment with intralymphatic GAD-alum has been shown to be tolerable and safe in persons with type 1 diabetes and has shown significant efficacy to preserve C-peptide with associated clinical benefit in individuals with the human leukocyte antigen DR3DQ2 haplotype. To further explore the feasibility and advantages of redosing with intralymphatic GAD-alum, six participants who had previously received active treatment with intralymphatic GAD-alum and carried HLA DR3-DQ2 received one additional intralymphatic dose of 4 μg GAD-alum in the pilot trial DIAGNODE-B. The participants also received 2000 U/day vitamin D (Calciferol) supplementation for two months, starting one month prior to the GAD-alum injection. During the 12-month follow-up, residual beta cell function was estimated with Mixed-Meal Tolerance Tests, and clinical and immune responses were observed. C-peptide decreased minimally, and most patients showed stable HbA1c and IDAA1c. The mean % TIR increased while the mean daily insulin dose decreased at month 12 compared to the baseline. Redosing with GAD-alum seems to be safe and tolerable, and may prolong the disease modification elicited by the original GAD-alum treatment.
Collapse
Affiliation(s)
- Rosaura Casas
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, 581 83 Linköping, Sweden;
| | - Andrea Tompa
- Department of Clinical Diagnostics, School of Health and Welfare, Jönköping University, 551 11 Jönköping, Sweden;
- Division of Medical Diagnostics, Department of Laboratory Medicine, Ryhov County Hospital, 551 85 Jönköping, Sweden
| | - Karin Åkesson
- Department of Pediatrics, Ryhov County Hospital, 551 85 Jönköping, Sweden;
| | | | - Anton Lindqvist
- Diamyd Medical AB, 111 56 Stockholm, Sweden; (P.F.T.); (A.L.)
| | - Johnny Ludvigsson
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, 581 83 Linköping, Sweden;
- Crown Princess Victoria Children’s Hospital, Linköping University, 581 85 Linköping, Sweden
| |
Collapse
|
2
|
Barra JM, Robino RA, Castro-Gutierrez R, Proia J, Russ HA, Ferreira LMR. Combinatorial genetic engineering strategy for immune protection of stem cell-derived beta cells by chimeric antigen receptor regulatory T cells. Cell Rep 2024; 43:114994. [PMID: 39561045 PMCID: PMC11659569 DOI: 10.1016/j.celrep.2024.114994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 10/07/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Abstract
Regenerative medicine is a rapidly expanding field harnessing human pluripotent stem cell (hPSC)-derived cells and tissues to treat many diseases, including type 1 diabetes. However, graft immune protection remains a key challenge. Chimeric antigen receptor (CAR) technology confers new specificities to effector T cells and immunosuppressive regulatory T cells (Tregs). One challenge in CAR design is identifying target molecules unique to the cells of interest. Here, we employ combinatorial genetic engineering to confer CAR-Treg-mediated localized immune protection to stem cell-derived cells. We engineered hPSCs to express truncated epidermal growth factor receptor (EGFRt), a biologically inert and generalizable target for CAR-Treg homing and activation, and generated CAR-Tregs recognizing EGFRt. Strikingly, CAR-Tregs suppressed innate and adaptive immune responses in vitro and prevented EGFRt-hPSC-derived pancreatic beta-like cell (sBC [stem cell-derived beta cell]) graft immune destruction in vivo. Collectively, we provide proof of concept that hPSCs and Tregs can be co-engineered to protect hPSC-derived cells from immune rejection upon transplantation.
Collapse
Affiliation(s)
- Jessie M Barra
- Diabetes Institute, University of Florida, Gainesville, FL 32610, USA; Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA
| | - Rob A Robino
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Roberto Castro-Gutierrez
- Diabetes Institute, University of Florida, Gainesville, FL 32610, USA; Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA
| | - James Proia
- Diabetes Institute, University of Florida, Gainesville, FL 32610, USA; Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA
| | - Holger A Russ
- Diabetes Institute, University of Florida, Gainesville, FL 32610, USA; Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA.
| | - Leonardo M R Ferreira
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
3
|
Leslie KA, Lekka C, Richardson SJ, Russell MA, Morgan NG. Regulation of STAT1 Signaling in Human Pancreatic β-Cells by the Lysine Deacetylase HDAC6: A New Therapeutic Opportunity in Type 1 Diabetes? Diabetes 2024; 73:1473-1485. [PMID: 38869827 DOI: 10.2337/db24-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 06/05/2024] [Indexed: 06/14/2024]
Abstract
Type 1 diabetes arises from the selective destruction of pancreatic β-cells by autoimmune mechanisms, and intracellular pathways driven by Janus kinase (JAK)-mediated phosphorylation of STAT isoforms (especially STAT1 and STAT2) are implicated as mediators of β-cell demise. Despite this, the molecular mechanisms that regulate JAK-STAT signaling in β-cells during the autoimmune attack remain only partially disclosed, and the factors acting to antagonize proinflammatory STAT1 signaling are uncertain. We have recently implicated signal regulatory protein α (SIRPα) in promoting β-cell viability in the face of ongoing islet autoimmunity and have now revealed that this protein controls the availability of a cytosolic lysine deacetylase, HDAC6, whose activity regulates the phosphorylation and activation of STAT1. We provide evidence that STAT1 serves as a substrate for HDAC6 in β-cells and that sequestration of HDAC6 by SIRPα in response to anti-inflammatory cytokines (e.g., IL-13) leads to increased STAT1 acetylation. This then impairs the ability of STAT1 to promote gene transcription in response to proinflammatory cytokines, including interferon-γ. We further found that SIRPα is lost from the β-cells of subjects with recent-onset type 1 diabetes under conditions when HDAC6 is retained and STAT1 levels are increased. On this basis, we report a previously unrecognized role for cytokine-induced regulation of STAT1 acetylation in the control of β-cell viability and propose that targeted inhibition of HDAC6 activity may represent a novel therapeutic modality to promote β-cell viability in the face of active islet autoimmunity. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Kaiyven Afi Leslie
- Islet Biology Group (IBEx), Exeter Centre of Excellence in Diabetes (EXCEED), Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, U.K
| | - Christiana Lekka
- Islet Biology Group (IBEx), Exeter Centre of Excellence in Diabetes (EXCEED), Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, U.K
| | - Sarah J Richardson
- Islet Biology Group (IBEx), Exeter Centre of Excellence in Diabetes (EXCEED), Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, U.K
| | - Mark A Russell
- Islet Biology Group (IBEx), Exeter Centre of Excellence in Diabetes (EXCEED), Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, U.K
| | | |
Collapse
|
4
|
Kado T, Tomimaru Y, Kobayashi S, Harada A, Sasaki K, Iwagami Y, Yamada D, Noda T, Takahashi H, Kita S, Shimomura I, Miyagawa S, Doki Y, Eguchi H. Skeletal Myoblast Cells Enhance the Function of Transplanted Islets in Diabetic Mice. J Diabetes Res 2024; 2024:5574968. [PMID: 38800586 PMCID: PMC11126349 DOI: 10.1155/2024/5574968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/05/2024] [Accepted: 04/23/2024] [Indexed: 05/29/2024] Open
Abstract
Islet transplantation (ITx) is an established and safe alternative to pancreas transplantation for type 1 diabetes mellitus (T1DM) patients. However, most ITx recipients lose insulin independence by 3 years after ITx due to early graft loss, such that multiple donors are required to achieve insulin independence. In the present study, we investigated whether skeletal myoblast cells could be beneficial for promoting angiogenesis and maintaining the differentiated phenotypes of islets. In vitro experiments showed that the myoblast cells secreted angiogenesis-related cytokines (vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), and stromal-derived factor-1α (SDF-1α)), contributed to maintenance of differentiated islet phenotypes, and enhanced islet cell insulin secretion capacity. To verify these findings in vivo, we transplanted islets alone or with myoblast cells under the kidney capsule of streptozotocin-induced diabetic mice. Compared with islets alone, the group bearing islets with myoblast cells had a significantly lower average blood glucose level. Histological examination revealed that transplants with islets plus myoblast cells were associated with a significantly larger insulin-positive area and significantly higher number of CD31-positive microvessels compared to islets alone. Furthermore, islets cotransplanted with myoblast cells showed JAK-STAT signaling activation. Our results suggest two possible mechanisms underlying enhancement of islet graft function with myoblast cells cotransplantation: "indirect effects" mediated by angiogenesis and "direct effects" of myoblast cells on islets via the JAK-STAT cascade. Overall, these findings suggest that skeletal myoblast cells enhance the function of transplanted islets, implying clinical potential for a novel ITx procedure involving myoblast cells for patients with diabetes.
Collapse
Affiliation(s)
- Takeshi Kado
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yoshito Tomimaru
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shogo Kobayashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Akima Harada
- Department of Cardiovascular Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kazuki Sasaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yoshifumi Iwagami
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Daisaku Yamada
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takehiro Noda
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hidenori Takahashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shunbun Kita
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Adipose Management, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
5
|
Tornai D, Mitchell M, McClain CJ, Dasarathy S, McCullough A, Radaeva S, Kroll-Desrosiers A, Lee J, Barton B, Szabo G. A novel score of IL-13 and age predicts 90-day mortality in severe alcohol-associated hepatitis: A multicenter plasma biomarker analysis. Hepatol Commun 2023; 7:e0296. [PMID: 37994498 PMCID: PMC10666984 DOI: 10.1097/hc9.0000000000000296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 08/15/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND Severe alcoholic hepatitis (AH) has a high short-term mortality rate. The MELD assesses disease severity and mortality; however, it is not specific for AH. We screened plasma samples from patients with severe AH for biomarkers of multiple pathological processes and identified predictors of short-term mortality. METHODS Plasma was collected at baseline from 85 patients with severe AH (MELD≥20, Maddrey's discriminant function≥32) enrolled in the Defeat Alcoholic Steatohepatitis clinical trial (investigating IL-1 receptor antagonist+pentoxifylline+zinc vs. methylprednisolone+placebo). Samples were analyzed for 43 biomarkers and the markers' association with 28- and 90-day mortalities was assessed. RESULTS Thirty-one (36.5%) patients died during the 90-day follow-up with similar ratios in the treatment groups. Eight biomarkers showed an association with mortality. IL-6, IL-22, interferon-α2, soluble TNF receptor 1, lipocalin-2, and α-fetoprotein levels were associated with 28-day mortality, while IL-6, IL-13, and endotoxin levels with 90-day mortality. In multivariable Cox regression, encephalopathy, lipocalin-2, and α-fetoprotein levels were independent predictors of 28-day mortality, and IL-6, IL-13, international normalized ratio levels, and age were independent predictors of 90-day mortality. The combination of IL-13 and age had superior performance in predicting 90-day mortality compared with MELD in the total cohort and the individual treatment groups. CONCLUSIONS We identified predictors of short-term mortality in a cohort exclusively involving patients with severe AH. We created a composite score of IL-13 and age that predicts 90-day mortality regardless of the treatment type with a performance superior to MELD in severe AH.
Collapse
Affiliation(s)
- David Tornai
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
- Department of Internal Medicine, Division of Gastroenterology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Mack Mitchell
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Craig J. McClain
- Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Srinivasan Dasarathy
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, Ohio, USA
- Department of Inflammation and Immunity, Lerner Research Institute of the Cleveland Clinic, Cleveland, Ohio, USA
| | - Arthur McCullough
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, Ohio, USA
- Department of Inflammation and Immunity, Lerner Research Institute of the Cleveland Clinic, Cleveland, Ohio, USA
| | - Svetlana Radaeva
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, Marylansd, USA
| | - Aimee Kroll-Desrosiers
- Department of Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- VA Central Western Massachusetts Healthcare System, Leeds, Massachusetts, USA
| | - JungAe Lee
- Department of Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Bruce Barton
- Department of Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Gyongyi Szabo
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Russell MA, Richardson SJ, Morgan NG. The role of the interferon/JAK-STAT axis in driving islet HLA-I hyperexpression in type 1 diabetes. Front Endocrinol (Lausanne) 2023; 14:1270325. [PMID: 37867531 PMCID: PMC10588626 DOI: 10.3389/fendo.2023.1270325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/06/2023] [Indexed: 10/24/2023] Open
Abstract
The hyperexpression of human leukocyte antigen class I (HLA-I) molecules on pancreatic beta-cells is widely accepted as a hallmark feature of type 1 diabetes pathogenesis. This response is important clinically since it may increase the visibility of beta-cells to autoreactive CD8+ T-cells, thereby accelerating disease progression. In this review, key factors which drive HLA-I hyperexpression will be explored, and their clinical significance examined. It is established that the presence of residual beta-cells is essential for HLA-I hyperexpression by islet cells at all stages of the disease. We suggest that the most likely drivers of this process are interferons released from beta-cells (type I or III interferon; possibly in response to viral infection) or those elaborated from influent, autoreactive immune cells (type II interferon). In both cases, Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) pathways will be activated to induce the downstream expression of interferon stimulated genes. A variety of models have highlighted that HLA-I expression is enhanced in beta-cells in response to interferons, and that STAT1, STAT2 and interferon regulatory factor 9 (IRF9) play key roles in mediating these effects (depending on the species of interferon involved). Importantly, STAT1 expression is elevated in the beta-cells of donors with recent-onset type I diabetes, and this correlates with HLA-I hyperexpression on an islet-by-islet basis. These responses can be replicated in vitro, and we consider that chronically elevated STAT1 may have a role in maintaining HLA-I hyperexpression. However, other data have highlighted that STAT2-IRF9 may also be critical to this process. Thus, a better understanding of how these factors regulate HLA-I under chronically stimulated conditions needs to be gathered. Finally, JAK inhibitors can target interferon signaling pathways to diminish HLA-I expression in mouse models. It seems probable that these agents may also be effective in patients; diminishing HLA-I hyperexpression on islets, reducing the visibility of beta-cells to the immune system and ultimately slowing disease progression. The first clinical trials of selective JAK inhibitors are underway, and the outcomes should have important implications for type 1 diabetes clinical management.
Collapse
Affiliation(s)
- Mark A. Russell
- Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, United Kingdom
| | | | | |
Collapse
|
7
|
Sionov RV, Ahdut-HaCohen R. A Supportive Role of Mesenchymal Stem Cells on Insulin-Producing Langerhans Islets with a Specific Emphasis on The Secretome. Biomedicines 2023; 11:2558. [PMID: 37761001 PMCID: PMC10527322 DOI: 10.3390/biomedicines11092558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/06/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Type 1 Diabetes (T1D) is a chronic autoimmune disease characterized by a gradual destruction of insulin-producing β-cells in the endocrine pancreas due to innate and specific immune responses, leading to impaired glucose homeostasis. T1D patients usually require regular insulin injections after meals to maintain normal serum glucose levels. In severe cases, pancreas or Langerhans islet transplantation can assist in reaching a sufficient β-mass to normalize glucose homeostasis. The latter procedure is limited because of low donor availability, high islet loss, and immune rejection. There is still a need to develop new technologies to improve islet survival and implantation and to keep the islets functional. Mesenchymal stem cells (MSCs) are multipotent non-hematopoietic progenitor cells with high plasticity that can support human pancreatic islet function both in vitro and in vivo and islet co-transplantation with MSCs is more effective than islet transplantation alone in attenuating diabetes progression. The beneficial effect of MSCs on islet function is due to a combined effect on angiogenesis, suppression of immune responses, and secretion of growth factors essential for islet survival and function. In this review, various aspects of MSCs related to islet function and diabetes are described.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- The Institute of Biomedical and Oral Research (IBOR), Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Ronit Ahdut-HaCohen
- Department of Medical Neurobiology, Institute of Medical Research, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel;
- Department of Science, The David Yellin Academic College of Education, Jerusalem 9103501, Israel
| |
Collapse
|
8
|
Ahmadi F, Lotfi AS, Navaei-Nigjeh M, Kadivar M. Trimetazidine Preconditioning Potentiates the Effect of Mesenchymal Stem Cells Secretome on the Preservation of Rat Pancreatic Islet Survival and Function In Vitro. Appl Biochem Biotechnol 2023; 195:4796-4817. [PMID: 37184724 DOI: 10.1007/s12010-023-04532-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 05/16/2023]
Abstract
Islet transplantation offers improved glycemic control in individuals with type 1 diabetes mellitus. However, in vitro islet culture is associated with islet apoptosis and eventually will lose their functionality prior to transplantation. In this study, we examined the effects of mesenchymal stem cells (MSCs) secretome preconditioned with diazoxide (DZ) and trimetazidine (TMZ) on rat islet cells during pre-transplant culture. With and without preconditioned hAD-MSCs' concentrated conditioned media (CCM) were added to the culture medium containing rat islets every 12 h for 24 and 48 h, after testing for selected cytokine concentrations (interleukin (IL)-4, IL-6, IL-13). Insulin content, glucose-stimulated insulin secretion, islet cell apoptosis, and mRNA expression of pro-apoptotic (BAX, BAK-1, and PUMA) and anti-apoptotic factors (BCL-2, BCL-xL, and XIAP) in rat islets were assessed after 24 and 48 h of culture. The protein level of IL-6 and IL-4 was significantly higher in TMZ-MSC-CM compared to MSC-non-CM. In rat isolated islets, normalized secreted insulin in the presence of 16.7 mM glucose was significantly higher in treated islet groups compared to control islets at both 24 and 48 h cultivation. Also, the percentage of apoptotic islet cells TMZ-MSC-CCM-treated islets was significantly lower compared to MSC-CM and MSC-CCM-treated islets in both 24 and 48 h cultivation. Consistent with the number of apoptotic cells, after 24 h culture, the expression of BCL-2 and BCL-xL genes in the control islets was lower than all treatment islet groups and in 48 h was lower than only TMZ-MSC-CM-treated islets. Also, the expression of the XIAP gene in control islets was significantly lower compared to the TMZ-MSC-CCM-treated islets at both at 24 and 48 h. In addition, mRNA level of the BAX gene in TMZ-MSC-CCM-treated islets was significantly lower compared to other groups at 48 h. Our findings revealed that TMZ proved to be more effective than DZ and could enhance the potential of hAD-MSCs-CM to improve the function and viability of islets prior to transplantation.
Collapse
Affiliation(s)
- Fariborz Ahmadi
- Department of Clinical Biochemistry, Tarbiat Modares University, Tehran, Iran
| | | | - Mona Navaei-Nigjeh
- Pharmaceutical Sciences Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mehdi Kadivar
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
9
|
Xu Y, Wei X, Li X, Chen Y, Mao X, Chen G, Liu C. Cadmium inhibits signal transducer and activator of transcription 6 leading to pancreatic β cell apoptosis. Endocr J 2022; 69:361-371. [PMID: 34719525 DOI: 10.1507/endocrj.ej21-0115] [Citation(s) in RCA: 1] [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: 11/23/2022] Open
Abstract
The toxic heavy metal cadmium has been proven to cause pancreatic dysfunction and lead to the development of DM. However, the underlying mechanisms have not been completely elucidated. Here, we investigated the effects of cadmium on the pancreatic β cell line MIN6 and explored the underlying mechanisms. The Cell Counting Kit-8 (CCK8) assay and flow cytometry were used to determine cell viability and apoptosis in MIN6 cells. The expression levels of signal transducer and activator of transcription 6 (STAT6) were assessed by western blotting. We further assessed the effects of cadmium on the function of pancreatic β cells under high glucose levels using enzyme-linked immunosorbent assay (ELISA) and western blotting. Insulin secretion and expression were decreased by cadmium in MIN6 cells. In addition, cadmium suppressed cell viability and promoted apoptosis of MIN6 cells, downregulated insulin secretion and genesis of MIN6 cells under high glucose conditions, while inhibiting STAT6. Furthermore, after treatment with IL-4, the activator of STAT6, the MIN6 cell viability suppression and apoptosis promotion effect caused by cadmium were blocked. In conclusion, cadmium inhibits pancreatic β cell MIN6 growth by regulating the activation of STAT6. Our findings reveal a new mechanism of cadmium toxicity in pancreatic β cells.
Collapse
Affiliation(s)
- Yijiao Xu
- Research Center of Endocrinology and Metabolic Diseases, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine of Nanjing University of Chinese Medicine, Qixia District, Nanjing, China, 210028
- Key Laboratory of State Administration of Traditional Chinese Medicine for TCM Syndrome & Treatment of Yingbing, Jiangsu Province Academy of Traditional Chinese Medicine, Qixia District, Nanjing, China, 210028
| | - Xiao Wei
- Research Center of Endocrinology and Metabolic Diseases, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine of Nanjing University of Chinese Medicine, Qixia District, Nanjing, China, 210028
- Key Laboratory of State Administration of Traditional Chinese Medicine for TCM Syndrome & Treatment of Yingbing, Jiangsu Province Academy of Traditional Chinese Medicine, Qixia District, Nanjing, China, 210028
| | - Xingjia Li
- Research Center of Endocrinology and Metabolic Diseases, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine of Nanjing University of Chinese Medicine, Qixia District, Nanjing, China, 210028
- Key Laboratory of State Administration of Traditional Chinese Medicine for TCM Syndrome & Treatment of Yingbing, Jiangsu Province Academy of Traditional Chinese Medicine, Qixia District, Nanjing, China, 210028
| | - Yu Chen
- Research Center of Endocrinology and Metabolic Diseases, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine of Nanjing University of Chinese Medicine, Qixia District, Nanjing, China, 210028
- Key Laboratory of State Administration of Traditional Chinese Medicine for TCM Syndrome & Treatment of Yingbing, Jiangsu Province Academy of Traditional Chinese Medicine, Qixia District, Nanjing, China, 210028
| | - Xiaodong Mao
- Research Center of Endocrinology and Metabolic Diseases, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine of Nanjing University of Chinese Medicine, Qixia District, Nanjing, China, 210028
- Key Laboratory of State Administration of Traditional Chinese Medicine for TCM Syndrome & Treatment of Yingbing, Jiangsu Province Academy of Traditional Chinese Medicine, Qixia District, Nanjing, China, 210028
| | - Guofang Chen
- Research Center of Endocrinology and Metabolic Diseases, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine of Nanjing University of Chinese Medicine, Qixia District, Nanjing, China, 210028
- Key Laboratory of State Administration of Traditional Chinese Medicine for TCM Syndrome & Treatment of Yingbing, Jiangsu Province Academy of Traditional Chinese Medicine, Qixia District, Nanjing, China, 210028
| | - Chao Liu
- Research Center of Endocrinology and Metabolic Diseases, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine of Nanjing University of Chinese Medicine, Qixia District, Nanjing, China, 210028
- Key Laboratory of State Administration of Traditional Chinese Medicine for TCM Syndrome & Treatment of Yingbing, Jiangsu Province Academy of Traditional Chinese Medicine, Qixia District, Nanjing, China, 210028
| |
Collapse
|
10
|
Secco B, Saitoski K, Drareni K, Soprani A, Pechberty S, Rachdi L, Venteclef N, Scharfmann R. Loss of Human Beta Cell Identity in a Reconstructed Omental Stromal Cell Environment. Cells 2022; 11:cells11060924. [PMID: 35326375 PMCID: PMC8946101 DOI: 10.3390/cells11060924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/01/2022] [Indexed: 11/30/2022] Open
Abstract
In human type 2 diabetes, adipose tissue plays an important role in disturbing glucose homeostasis by secreting factors that affect the function of cells and tissues throughout the body, including insulin-producing pancreatic beta cells. We aimed here at studying the paracrine effect of stromal cells isolated from subcutaneous and omental adipose tissue on human beta cells. We developed an in vitro model wherein the functional human beta cell line EndoC-βH1 was treated with conditioned media from human adipose tissues. By using RNA-sequencing and western blotting, we determined that a conditioned medium derived from omental stromal cells stimulates several pathways, such as STAT, SMAD and RELA, in EndoC-βH1 cells. We also observed that upon treatment, the expression of beta cell markers decreased while dedifferentiation markers increased. Loss-of-function experiments that efficiently blocked specific signaling pathways did not reverse dedifferentiation, suggesting the implication of more than one pathway in this regulatory process. Taken together, we demonstrate that soluble factors derived from stromal cells isolated from human omental adipose tissue signal human beta cells and modulate their identity.
Collapse
Affiliation(s)
- Blandine Secco
- Institut Cochin, Université de Paris, INSERM U1016, CNRS UMR 8104, 75014 Paris, France; (B.S.); (K.S.); (S.P.); (L.R.)
| | - Kevin Saitoski
- Institut Cochin, Université de Paris, INSERM U1016, CNRS UMR 8104, 75014 Paris, France; (B.S.); (K.S.); (S.P.); (L.R.)
| | - Karima Drareni
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Université de Paris, 75006 Paris, France; (K.D.); (A.S.); (N.V.)
| | - Antoine Soprani
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Université de Paris, 75006 Paris, France; (K.D.); (A.S.); (N.V.)
- Clinique Geoffroy Saint-Hilaire, Ramsey General de Santé, 75005 Paris, France
| | - Severine Pechberty
- Institut Cochin, Université de Paris, INSERM U1016, CNRS UMR 8104, 75014 Paris, France; (B.S.); (K.S.); (S.P.); (L.R.)
| | - Latif Rachdi
- Institut Cochin, Université de Paris, INSERM U1016, CNRS UMR 8104, 75014 Paris, France; (B.S.); (K.S.); (S.P.); (L.R.)
| | - Nicolas Venteclef
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Université de Paris, 75006 Paris, France; (K.D.); (A.S.); (N.V.)
| | - Raphaël Scharfmann
- Institut Cochin, Université de Paris, INSERM U1016, CNRS UMR 8104, 75014 Paris, France; (B.S.); (K.S.); (S.P.); (L.R.)
- Correspondence: ; Tel.: +(33)-1-76-53-55-68
| |
Collapse
|
11
|
Chakrabarti S, Dasgupta S, Deb A. Tumor infiltrating lymphocytes in head and neck squamous cell carcinoma – Evaluation and clinical impact. J Cancer Res Ther 2022; 18:49-54. [DOI: 10.4103/jcrt.jcrt_830_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
12
|
Leslie KA, Richardson SJ, Russell MA, Morgan NG. Expression of CD47 in the pancreatic β-cells of people with recent-onset type 1 diabetes varies according to disease endotype. Diabet Med 2021; 38:e14724. [PMID: 34654058 DOI: 10.1111/dme.14724] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/13/2021] [Indexed: 02/06/2023]
Abstract
AIMS We are studying the dialogue between β-cells and the immune system in type 1 diabetes and have identified a cell surface receptor, signal regulatory protein-alpha (SIRPα) as an important component in the regulation of β-cell survival. SIRPα interacts with another protein, CD47, to mediate signalling. In the present work, we have studied the expression and role of CD47 in human islet cells in type 1 diabetes. METHODS Clonal EndoC-βH1 cells were employed for functional studies. Cells were exposed to pro-inflammatory cytokines and their viability monitored by flow cytometry after staining with propidium iodide. Targeted knockdown of CD47 or SIRPα was achieved with small interference RNA molecules and the expression of relevant proteins studied by Western blotting or immunocytochemistry. Human pancreas sections were selected from the Exeter Archival Diabetes Biobank and used to examine the expression of CD47 by immunofluorescence labelling. Image analysis was employed to quantify expression. RESULTS CD47 is abundantly expressed in both α and β cells in human pancreas. In type 1 diabetes, the levels of CD47 are increased in α cells across all age groups, whereas the expression in β-cells varies according to disease endotype. Knockdown of either CD47 or SIRPα in EndoC-βH1 cells resulted in a loss of viability. CONCLUSIONS We conclude that the CD47 plays a previously unrecognised role in the regulation of β-cell viability. This system is dysregulated in type 1 diabetes suggesting that it may be targeted therapeutically to slow disease progression.
Collapse
Affiliation(s)
- Kaiyven Afi Leslie
- Islet Biology Group (IBEx), Exeter Centre of Excellence in Diabetes (EXCEED), University of Exeter College of Medicine and Health, Exeter, UK
| | - Sarah J Richardson
- Islet Biology Group (IBEx), Exeter Centre of Excellence in Diabetes (EXCEED), University of Exeter College of Medicine and Health, Exeter, UK
| | - Mark A Russell
- Islet Biology Group (IBEx), Exeter Centre of Excellence in Diabetes (EXCEED), University of Exeter College of Medicine and Health, Exeter, UK
| | - Noel G Morgan
- Islet Biology Group (IBEx), Exeter Centre of Excellence in Diabetes (EXCEED), University of Exeter College of Medicine and Health, Exeter, UK
| |
Collapse
|
13
|
Liu M, Wang Z, Feng D, Shang Y, Li X, Liu J, Li C, Yang Z. An Insulin-Inspired Supramolecular Hydrogel for Prevention of Type 1 Diabetes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003599. [PMID: 34026440 PMCID: PMC8132061 DOI: 10.1002/advs.202003599] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 01/11/2021] [Indexed: 05/10/2023]
Abstract
Supramolecular peptide hydrogel has shown promising potential in vaccine development largely because of its ability to function both as antigen depot and immune adjuvant. Nap-GdFdFdY, a tetrapeptide hydrogel that has been previously reported to exhibit adjuvant effect, is inadvertently found to contain conserved peptide sequence for insulin, proinsulin, and glutamic acid decarboxylase, 3 major autoantigens for the autoimmune type 1 diabetes (T1D). At present, despite being managed clinically with insulin replacement therapy, T1D remains a major health threat with rapidly increasing incidences, especially in children and young adults, and antigen-specific immune tolerance induction has been proposed as a feasible approach to prevent or delay T1D progression at an early stage. Here, it is reported that innoculation of Nap-GdFdFdY leads to complete protection of nonobese diabetic (NOD) mice from T1D development till the age of 36 weeks. Better maintenance of pancreatic islet morphology with minimal immune cell infiltration is also observed from mice exposed to Nap-GdFdFdY. This beneficial impact is mainly due to its facilitative role on enhancing peripheral T regulatory cell (Treg) population, shown as increased splenic Treg percentage, and function, demonstrated by maintenance of circulating TGF-β1 level. Serum cytokine microarray data further implicate a "buffering" role of Nap-GdFdFdY on systemic inflammatory tone in NOD mice. Thus, with its versatility, applicability, and excellent potency, Nap-GdFdFdY is posited as a novel therapeutic intervention for T1D.
Collapse
Affiliation(s)
- Mohan Liu
- Tianjin Key Laboratory of Biomedical MaterialsBiomedical Barriers Research CentreInstitute of Biomedical EngineeringChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300192P. R. China
| | - Zhongyan Wang
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyCollaborative Innovation Centre of Chemical Science and Engineeringand National Institute of Functional MaterialsNankai UniversityTianjin300071P. R. China
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear MedicineInstitute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300192P. R. China
| | - Dandan Feng
- Tianjin Key Laboratory of Biomedical MaterialsBiomedical Barriers Research CentreInstitute of Biomedical EngineeringChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300192P. R. China
| | - Yuna Shang
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyCollaborative Innovation Centre of Chemical Science and Engineeringand National Institute of Functional MaterialsNankai UniversityTianjin300071P. R. China
| | - Xinxin Li
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyCollaborative Innovation Centre of Chemical Science and Engineeringand National Institute of Functional MaterialsNankai UniversityTianjin300071P. R. China
| | - Jianfeng Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear MedicineInstitute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300192P. R. China
| | - Chen Li
- Tianjin Key Laboratory of Biomedical MaterialsBiomedical Barriers Research CentreInstitute of Biomedical EngineeringChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300192P. R. China
| | - Zhimou Yang
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyCollaborative Innovation Centre of Chemical Science and Engineeringand National Institute of Functional MaterialsNankai UniversityTianjin300071P. R. China
- Jiangsu Center for the Collaboration and Innovation of Cancer BiotherapyCancer InstituteXuzhou Medical UniversityXuzhouJiangsu221004P. R. China
| |
Collapse
|
14
|
Francisco D, Wang Y, Conway M, Hurbon AN, Dy ABC, Addison KJ, Chu HW, Voelker DR, Ledford JG, Kraft M. Surfactant Protein-A Protects against IL-13-Induced Inflammation in Asthma. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:2829-2839. [PMID: 32245819 PMCID: PMC7304346 DOI: 10.4049/jimmunol.1901227] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 03/09/2020] [Indexed: 11/19/2022]
Abstract
The lung surfactant proteins are recognized as critical not only for their role in lowering lung surface tension but also in innate host defense. Reports have shown that some asthmatic patients have decreased levels of one member of this protein family in particular, surfactant protein-A (SP-A). Our studies set out to determine the contribution of SP-A to the response of a key effector cytokine in asthma, IL-13. Our studies employ both animal models sufficient and deficient in SP-A challenged with IL-13 and primary epithelial cells from participants with asthma that are exogenously treated with SP-A in the context of IL-13 challenge. The inflammatory response and mucin production were assessed in both model systems. As compared with WT mice, we show that the activity of IL-13 is dramatically augmented in SP-A-/- mice, which have significantly increased neutrophil and eosinophil recruitment, mucin production and asthma-associated cytokines in the bronchoalveolar lavage fluid. In parallel, we show asthma-associated factors are attenuated in human cells from asthma subjects when exogenous SP-A is added during IL-13 challenge. Although many of these phenotypes have previously been associated with STAT6 signaling, SP-A inhibited IL-13-induced STAT3 phosphorylation in mice and in human epithelial cells while having little effect on STAT6 phosphorylation. In addition, when either STAT3 or IL-6 were inhibited in mice, the phenotypes observed in SP-A-/- mice were significantly attenuated. These studies suggest a novel mechanism for SP-A in asthma as a modulator of IL-13-induced inflammation via mediating downstream IL-6/STAT3 signaling.
Collapse
Affiliation(s)
- Dave Francisco
- Department of Medicine, University of Arizona, Tucson, AZ 85719
- Asthma and Airway Disease Research Center, Tucson, AZ 85724
- Department of Medicine, Duke University, Durham, NC 27707
| | - Ying Wang
- Department of Medicine, University of Arizona, Tucson, AZ 85719
- Asthma and Airway Disease Research Center, Tucson, AZ 85724
- Department of Medicine, Duke University, Durham, NC 27707
| | - Michelle Conway
- Department of Medicine, University of Arizona, Tucson, AZ 85719
| | | | - Alane B C Dy
- Asthma and Airway Disease Research Center, Tucson, AZ 85724
| | - Kenneth J Addison
- Asthma and Airway Disease Research Center, Tucson, AZ 85724
- Department of Medicine, Duke University, Durham, NC 27707
| | - Hong W Chu
- Department of Medicine, National Jewish Health, Denver, CO 80206; and
| | - Dennis R Voelker
- Department of Medicine, National Jewish Health, Denver, CO 80206; and
| | - Julie G Ledford
- Asthma and Airway Disease Research Center, Tucson, AZ 85724;
- Department of Medicine, Duke University, Durham, NC 27707
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85719
| | - Monica Kraft
- Department of Medicine, University of Arizona, Tucson, AZ 85719;
- Asthma and Airway Disease Research Center, Tucson, AZ 85724
- Department of Medicine, Duke University, Durham, NC 27707
| |
Collapse
|
15
|
Rajendran S, Graef M, Chu T, von Herrath M. IL-4R is expressed on alpha and beta cells of human pancreata. Clin Immunol 2020; 214:108394. [PMID: 32224156 DOI: 10.1016/j.clim.2020.108394] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 01/01/2023]
Affiliation(s)
- Sakthi Rajendran
- La Jolla Institute for Immunology, La Jolla, CA, United States of America
| | - Madeleine Graef
- La Jolla Institute for Immunology, La Jolla, CA, United States of America
| | - Tiffany Chu
- La Jolla Institute for Immunology, La Jolla, CA, United States of America
| | | |
Collapse
|
16
|
Colli ML, Szymczak F, Eizirik DL. Molecular Footprints of the Immune Assault on Pancreatic Beta Cells in Type 1 Diabetes. Front Endocrinol (Lausanne) 2020; 11:568446. [PMID: 33042023 PMCID: PMC7522353 DOI: 10.3389/fendo.2020.568446] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/17/2020] [Indexed: 12/25/2022] Open
Abstract
Type 1 diabetes (T1D) is a chronic disease caused by the selective destruction of the insulin-producing pancreatic beta cells by infiltrating immune cells. We presently evaluated the transcriptomic signature observed in beta cells in early T1D and compared it with the signatures observed following in vitro exposure of human islets to inflammatory or metabolic stresses, with the aim of identifying "footprints" of the immune assault in the target beta cells. We detected similarities between the beta cell signatures induced by cytokines present at different moments of the disease, i.e., interferon-α (early disease) and interleukin-1β plus interferon-γ (later stages) and the beta cells from T1D patients, identifying biological process and signaling pathways activated during early and late stages of the disease. Among the first responses triggered on beta cells was an enrichment in antiviral responses, pattern recognition receptors activation, protein modification and MHC class I antigen presentation. During putative later stages of insulitis the processes were dominated by T-cell recruitment and activation and attempts of beta cells to defend themselves through the activation of anti-inflammatory pathways (i.e., IL10, IL4/13) and immune check-point proteins (i.e., PDL1 and HLA-E). Finally, we mined the beta cell signature in islets from T1D patients using the Connectivity Map, a large database of chemical compounds/drugs, and identified interesting candidates to potentially revert the effects of insulitis on beta cells.
Collapse
Affiliation(s)
- Maikel L. Colli
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), Brussels, Belgium
- *Correspondence: Maikel L. Colli
| | - Florian Szymczak
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Decio L. Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Welbio, Medical Faculty, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Indiana Biosciences Research Institute, Indianapolis, IN, United States
| |
Collapse
|
17
|
IL-6 is present in beta and alpha cells in human pancreatic islets: Expression is reduced in subjects with type 1 diabetes. Clin Immunol 2019; 211:108320. [PMID: 31809899 DOI: 10.1016/j.clim.2019.108320] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/27/2019] [Accepted: 11/29/2019] [Indexed: 01/03/2023]
Abstract
IL-6 is a pro-inflammatory cytokine upregulated in some autoimmune diseases. The role of IL-6 in the development of type 1 diabetes (T1D) is unclear. Clinical studies are investigating whether tocilizumab (anti-IL-6 receptor) can help preserve beta cell function in patients recently diagnosed with T1D. However, in some rodent models and isolated human islets, IL-6 has been found to have a protective role for beta cells by reducing oxidative stress. Hence, we systematically investigated local tissue expression of IL-6 in human pancreas from non-diabetic, auto-antibody positive donors and donors with T1D and T2D. IL-6 was constitutively expressed by beta and alpha cells regardless of the disease state. However, expression of IL-6 was highly reduced in insulin-deficient islets of donors with T1D, and the expression was then mostly restricted to alpha cells. Our findings suggest that the implication of IL-6 in T1D pathogenesis might be more complex than previously assumed.
Collapse
|
18
|
Hu L, Li L, Zhang H, Li Q, Jiang S, Qiu J, Sun J, Dong J. Inhibition of airway remodeling and inflammatory response by Icariin in asthma. Altern Ther Health Med 2019; 19:316. [PMID: 31744482 PMCID: PMC6862818 DOI: 10.1186/s12906-019-2743-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 11/04/2019] [Indexed: 11/10/2022]
Abstract
BACKGROUND Icariin (ICA) is the major active ingredient extracted from Chinese herbal medicine Epimedium, which has the effects of improving cardiovascular function, inducing tumor cell differentiation and increasing bone formation. It is still rarely reported that ICA can exert its therapeutic potential in asthma via anti-airway remodeling. The point of the study was to estimate the role of ICA in anti-. airway remodeling and its possible mechanism of action in a mouse ovalbumin. (OVA)-induced asthma model. METHODS Hematoxylin and Eosin Staining were performed for measuring airway remodeling related indicators. ELISA, Western blot and Immunohistochemistr-. y (IHC) were used for analyzing the level of protein. RT-PCR was used for analyzing the level of mRNA. RESULTS On days 1 and 8, mice were sensitized to OVA by intraperitoneal injection. From day 16 to day 43, previously sensitized mice were exposed to OVA once daily by nebulizer. Interventions were performed orally with ICA (ICA low, medium and high dose groups) or dexamethasone 1 h prior to each OVA exposure. ICA improves pulmonary function, attenuates pulmonary inflammation and airway remodeling in mice exposed to OVA. Histological and Western blot analysis of the lungs show that ICA suppressed transforming growth factor beta 1 and vascular endothelial growth factor expression. Increase in interleukin 13 and endothelin-1 in serum and bronchoalveolar lavage fluid in OVA-induced asthmatic mice are also decreased by ICA. ICA attenuates airway smooth muscle cell proliferation, as well as key factors in the MAPK/Erk pathway. CONCLUSIONS The fact that ICA can alleviate OVA-induced asthma at least partly through inhibition of ASMC proliferation via MAPK/Erk pathway provides a solid theoretical basis for ICA as a replacement therapy for asthma. These data reveal the underlying reasons of the use of ICA-rich herbs in Traditional Chinese Medicine to achieve good results in treating asthma.
Collapse
|
19
|
Sharba S, Navabi N, Padra M, Persson JA, Quintana-Hayashi MP, Gustafsson JK, Szeponik L, Venkatakrishnan V, Sjöling Å, Nilsson S, Quiding-Järbrink M, Johansson MEV, Linden SK. Interleukin 4 induces rapid mucin transport, increases mucus thickness and quality and decreases colitis and Citrobacter rodentium in contact with epithelial cells. Virulence 2019; 10:97-117. [PMID: 30665337 PMCID: PMC6363059 DOI: 10.1080/21505594.2019.1573050] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Citrobacter rodentium infection is a murine model for pathogenic intestinal Escherichia coli infection. C. rodentium infection causes an initial decrease in mucus layer thickness, followed by an increase during clearance. We aimed to identify the cause of these changes and to utilize this naturally occurring mucus stimulus to decrease pathogen impact and inflammation. We identified that mucin production and speed of transport from Golgi to secretory vesicles at the apical surface increased concomitantly with increased mucus thickness. Of the cytokines differentially expressed during increased mucus thickness, IFN-γ and TNF-α decreased the mucin production and transport speed, whereas IL-4, IL-13, C. rodentium and E. coli enhanced these aspects. IFN-γ and TNF-α treatment in combination with C. rodentium and pathogenic E. coli infection negatively affected mucus parameters in vitro, which was relieved by IL-4 treatment. The effect of IL-4 was more pronounced than that of IL-13, and in wild type mice, only IL-4 was present. Increased expression of Il-4, Il-4-receptor α, Stat6 and Spdef during clearance indicate that this pathway contributes to the increase in mucin production. In vivo IL-4 administration initiated 10 days after infection increased mucus thickness and quality and decreased colitis and pathogen contact with the epithelium. Thus, during clearance of infection, the concomitant increase in IL-4 protects and maintains goblet cell function against the increasing levels of TNF-α and IFN-γ. Furthermore, IL-4 affects intestinal mucus production, pathogen contact with the epithelium and colitis. IL-4 treatment may thus have therapeutic benefits for mucosal healing.
Collapse
Affiliation(s)
- S Sharba
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - N Navabi
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - M Padra
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - J A Persson
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - M P Quintana-Hayashi
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - J K Gustafsson
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - L Szeponik
- b Department of Microbiology and Immunology , Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - V Venkatakrishnan
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - Å Sjöling
- c Department of Microbiology, Tumor and Cell Biology , Karolinska Institutet , Stockholm , Sweden
| | - S Nilsson
- d Department of Pathology & Genetics, Sahlgrenska Academy , University of Gothenburg , Sweden.,e Department of Mathematical Sciences , Chalmer University of Technology , Gothenburg , Sweden
| | - M Quiding-Järbrink
- b Department of Microbiology and Immunology , Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - M E V Johansson
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - S K Linden
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| |
Collapse
|
20
|
Leslie KA, Russell MA, Taniguchi K, Richardson SJ, Morgan NG. The transcription factor STAT6 plays a critical role in promoting beta cell viability and is depleted in islets of individuals with type 1 diabetes. Diabetologia 2019; 62:87-98. [PMID: 30338340 PMCID: PMC6290857 DOI: 10.1007/s00125-018-4750-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/14/2018] [Indexed: 12/17/2022]
Abstract
AIMS/HYPOTHESIS In type 1 diabetes, selective beta cell loss occurs within the inflamed milieu of insulitic islets. This milieu is generated via the enhanced secretion of proinflammatory cytokines and by the loss of anti-inflammatory molecules such as IL-4 and IL-13. While the actions of proinflammatory cytokines have been well-studied in beta cells, the effects of their anti-inflammatory counterparts have received relatively little attention and we have addressed this. METHODS Clonal beta cells, isolated human islets and pancreas sections from control individuals and those with type 1 diabetes were employed. Gene expression was measured using targeted gene arrays and by quantitative RT-PCR. Protein expression was monitored in cell extracts by western blotting and in tissue sections by immunocytochemistry. Target proteins were knocked down selectively with interference RNA. RESULTS Cytoprotection achieved with IL-4 and IL-13 is mediated by the early activation of signal transducer and activator of transcription 6 (STAT6) in beta cells, leading to the upregulation of anti-apoptotic proteins, including myeloid leukaemia-1 (MCL-1) and B cell lymphoma-extra large (BCLXL). We also report the induction of signal regulatory protein-α (SIRPα), and find that knockdown of SIRPα is associated with reduced beta cell viability. These anti-apoptotic proteins and their attendant cytoprotective effects are lost following siRNA-mediated knockdown of STAT6 in beta cells. Importantly, analysis of human pancreas sections revealed that STAT6 is markedly depleted in the beta cells of individuals with type 1 diabetes, implying the loss of cytoprotective responses. CONCLUSIONS/INTERPRETATION Selective loss of STAT6 may contribute to beta cell demise during the progression of type 1 diabetes.
Collapse
Affiliation(s)
- Kaiyven A Leslie
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, RILD Building (Level 4), Barrack Road, Exeter, EX2 5DW, UK
| | - Mark A Russell
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, RILD Building (Level 4), Barrack Road, Exeter, EX2 5DW, UK.
| | - Kazuto Taniguchi
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, RILD Building (Level 4), Barrack Road, Exeter, EX2 5DW, UK
| | - Sarah J Richardson
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, RILD Building (Level 4), Barrack Road, Exeter, EX2 5DW, UK
| | - Noel G Morgan
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, RILD Building (Level 4), Barrack Road, Exeter, EX2 5DW, UK.
| |
Collapse
|
21
|
Mao YM, Zhao CN, Leng J, Leng RX, Ye DQ, Zheng SG, Pan HF. Interleukin-13: A promising therapeutic target for autoimmune disease. Cytokine Growth Factor Rev 2018; 45:9-23. [PMID: 30581068 DOI: 10.1016/j.cytogfr.2018.12.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 12/11/2018] [Indexed: 12/16/2022]
Abstract
Interleukin-13 (IL-13) was previously thought to be a redundant presence of IL-4, but in recent years its role in immunity, inflammation, fibrosis, and allergic diseases has become increasingly prominent. IL-13 can regulate several subtypes of T helper (Th) cells and affect their transformation, including Th1, Th2, T17, etc., thus it may play an important role in immune system. Previous studies have revealed that IL-13 is implicated in the pathogenesis of autoimmune diseases, such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), systemic sclerosis (SSc), ulcerative colitis (UC), type 1 diabetes (T1D), sjogren's syndrome (SS), etc. In this review, we will briefly discuss the biological features of IL-13 and summarize recent advances in the role of IL-13 in the development and pathogenesis of autoimmune diseases. This information may provide new perspectives and suggestions for the selection of therapeutic targets for autoimmune diseases.
Collapse
Affiliation(s)
- Yan-Mei Mao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, 230032, China
| | - Chan-Na Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, 230032, China
| | - Jing Leng
- Anhui Academy of Medical Sciences, 15 Yonghong Road, Hefei, Anhui, China
| | - Rui-Xue Leng
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, 230032, China
| | - Dong-Qing Ye
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, 230032, China
| | - Song Guo Zheng
- Division of Rheumatology, Penn State College of Medicine, Hershey, PA 17033, USA.
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, 230032, China.
| |
Collapse
|
22
|
Tavira B, Barcenilla H, Wahlberg J, Achenbach P, Ludvigsson J, Casas R. Intralymphatic Glutamic Acid Decarboxylase-Alum Administration Induced Th2-Like-Specific Immunomodulation in Responder Patients: A Pilot Clinical Trial in Type 1 Diabetes. J Diabetes Res 2018; 2018:9391845. [PMID: 30009185 PMCID: PMC5994289 DOI: 10.1155/2018/9391845] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/06/2018] [Accepted: 04/24/2018] [Indexed: 01/10/2023] Open
Abstract
GAD-alum given into lymph nodes to type 1 diabetes patients participating in an open-label pilot trial resulted in preservation of C-peptide similar to promising results from other trials. Here, we compared the immunomodulatory effect of giving GAD-alum directly into lymph nodes versus that induced by subcutaneous administration. Samples from T1D patients (n = 6) who received 4 μg GAD-alum into lymph nodes (LNs), followed by two booster injections one month apart, and from patients (n = 6) who received two subcutaneous injections (SC) (20 μg) given one month apart were compared. GADA, IA-2A, GADA subclasses, IgE, GAD65-induced cytokines, PBMC proliferation, and T cell markers were analyzed. Lower doses of GAD-alum into LN induced higher GADA levels than SC injections and reduced proliferation and IgG1 GADA subclass, while enhancing IgG2, IgG3, and IgG4. The cytokine profile was dominated by the Th2-associated cytokine IL-13, and GAD65 stimulation induced activated CD4 T cells. Patients responding clinically best account for most of the immunological changes. In contrast, SC treatment resulted in predominant IgG1, predominant IFN-γ, higher proliferation, and activated CD4 and CD8 cells. Patients from the LN group with best metabolic outcome seemed to have common immune correlates related to the treatment. This trial is registered with DIAGNODE (NCT02352974, clinicaltrials.gov) and DIABGAD (NCT01785108, clinicaltrials.gov).
Collapse
Affiliation(s)
- Beatriz Tavira
- Division of Pediatrics, Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Hugo Barcenilla
- Division of Pediatrics, Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Jeannette Wahlberg
- Department of Endocrinology and Department of Medical and Health Sciences and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Peter Achenbach
- Institute of Diabetes Research, Helmholtz Zentrum München, and Forschergruppe Diabetes, Klinikum Rechts der Isar, Technische Universität München, München, Germany
| | - Johnny Ludvigsson
- Division of Pediatrics, Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
- Crown Princess Victoria Children's Hospital, Region Östergötland, Linköping, Sweden
| | - Rosaura Casas
- Division of Pediatrics, Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
23
|
Sadreev II, Chen MZQ, Umezawa Y, Biktashev VN, Kemper C, Salakhieva DV, Welsh GI, Kotov NV. The competitive nature of signal transducer and activator of transcription complex formation drives phenotype switching of T cells. Immunology 2017; 153:488-501. [PMID: 29030870 DOI: 10.1111/imm.12851] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 09/16/2017] [Accepted: 10/03/2017] [Indexed: 12/12/2022] Open
Abstract
Signal transducers and activators of transcription (STATs) are key molecular determinants of T-cell fate and effector function. Several inflammatory diseases are characterized by an altered balance of T-cell phenotypes and cytokine secretion. STATs, therefore, represent viable therapeutic targets in numerous pathologies. However, the underlying mechanisms by which the same STAT proteins regulate both the development of different T-cell phenotypes and their plasticity during changes in extracellular conditions remain unclear. In this study, we investigated the STAT-mediated regulation of T-cell phenotype formation and plasticity using mathematical modelling and experimental data for intracellular STAT signalling proteins. The close fit of our model predictions to the experimental data allows us to propose a potential mechanism for T-cell switching. According to this mechanism, T-cell phenotype switching is the result of the relative redistribution of STAT dimer complexes caused by the extracellular cytokine-dependent STAT competition effects. The developed model predicts that the balance between the intracellular STAT species defines the amount of the produced cytokines and thereby T-cell phenotypes. The model predictions are consistent with the experimentally observed interferon-γ to interleukin-10 switching that regulates human T helper type 1/type 1 regulatory T-cell responses. The proposed model is applicable to a number of STAT signalling circuits.
Collapse
Affiliation(s)
- Ildar I Sadreev
- Centre for Systems, Dynamics and Control, College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, UK
| | - Michael Z Q Chen
- School of Automation, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
| | - Yoshinori Umezawa
- Department of Dermatology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Vadim N Biktashev
- Centre for Systems, Dynamics and Control, College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, UK
| | - Claudia Kemper
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung and Blood Institute, Bethesda, MD, USA.,Division of Transplant Immunology and Mucosal Biology, King's College London, London, UK.,Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Diana V Salakhieva
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia
| | - Gavin I Welsh
- Bristol Renal, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Nikolay V Kotov
- Biophysics & Bionics Laboratory, Institute of Physics, Kazan (Volga Region) Federal University, Kazan, Russia
| |
Collapse
|
24
|
Kurian SM, Ferreri K, Wang CH, Todorov I, Al-Abdullah IH, Rawson J, Mullen Y, Salomon DR, Kandeel F. Gene expression signature predicts human islet integrity and transplant functionality in diabetic mice. PLoS One 2017; 12:e0185331. [PMID: 28968432 PMCID: PMC5624587 DOI: 10.1371/journal.pone.0185331] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 09/11/2017] [Indexed: 11/18/2022] Open
Abstract
There is growing evidence that transplantation of cadaveric human islets is an effective therapy for type 1 diabetes. However, gauging the suitability of islet samples for clinical use remains a challenge. We hypothesized that islet quality is reflected in the expression of specific genes. Therefore, gene expression in 59 human islet preparations was analyzed and correlated with diabetes reversal after transplantation in diabetic mice. Analysis yielded 262 differentially expressed probesets, which together predict islet quality with 83% accuracy. Pathway analysis revealed that failing islet preparations activated inflammatory pathways, while functional islets showed increased regeneration pathway gene expression. Gene expression associated with apoptosis and oxygen consumption showed little overlap with each other or with the 262 probeset classifier, indicating that the three tests are measuring different aspects of islet cell biology. A subset of 36 probesets surpassed the predictive accuracy of the entire set for reversal of diabetes, and was further reduced by logistic regression to sets of 14 and 5 without losing accuracy. These genes were further validated with an independent cohort of 16 samples. We believe this limited number of gene classifiers in combination with other tests may provide complementary verification of islet quality prior to their clinical use.
Collapse
Affiliation(s)
- Sunil M. Kurian
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - Kevin Ferreri
- Department of Translational Research and Cellular Therapeutics, Diabetes, and Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Chia-Hao Wang
- Department of Translational Research and Cellular Therapeutics, Diabetes, and Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Ivan Todorov
- Department of Translational Research and Cellular Therapeutics, Diabetes, and Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Ismail H. Al-Abdullah
- Department of Translational Research and Cellular Therapeutics, Diabetes, and Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Jeffrey Rawson
- Department of Translational Research and Cellular Therapeutics, Diabetes, and Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Yoko Mullen
- Department of Translational Research and Cellular Therapeutics, Diabetes, and Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Daniel R. Salomon
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - Fouad Kandeel
- Department of Translational Research and Cellular Therapeutics, Diabetes, and Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
- * E-mail:
| |
Collapse
|
25
|
Scharfmann R, Didiesheim M, Richards P, Chandra V, Oshima M, Albagli O. Mass production of functional human pancreatic β-cells: why and how? Diabetes Obes Metab 2016; 18 Suppl 1:128-36. [PMID: 27615142 DOI: 10.1111/dom.12728] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 05/17/2016] [Indexed: 12/17/2022]
Abstract
Diabetes (either type 1 or type 2) is due to insufficient functional β-cell mass. Research has, therefore, aimed to discover new ways to maintain or increase either β-cell mass or function. For this purpose, rodents have mainly been used as model systems and a large number of discoveries have been made. Meanwhile, although we have learned that rodent models represent powerful systems to model β-cell development, function and destruction, we realize that there are limitations when attempting to transfer the data to what is occurring in humans. Indeed, while human β-cells share many similarities with rodent β-cells, they also differ on a number of important parameters. In this context, developing ways to study human β-cell development, function and death represents an important challenge. This review will describe recent data on the development and use of convenient sources of human β-cells that should be useful tools to discover new ways to modulate functional β-cell mass in humans.
Collapse
Affiliation(s)
- R Scharfmann
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France.
| | - M Didiesheim
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - P Richards
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - V Chandra
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - M Oshima
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - O Albagli
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| |
Collapse
|
26
|
Tseng CC, Chang SJ, Tsai WC, Ou TT, Wu CC, Sung WY, Hsieh MC, Yen JH. Reduced incidence of Type 1 diabetes and Type 2 diabetes in systemic sclerosis: A nationwide cohort study. Joint Bone Spine 2016; 83:307-13. [DOI: 10.1016/j.jbspin.2015.06.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 06/21/2015] [Indexed: 10/22/2022]
|
27
|
Baltrusch S. Mitochondrial network regulation and its potential interference with inflammatory signals in pancreatic beta cells. Diabetologia 2016; 59:683-7. [PMID: 26873508 DOI: 10.1007/s00125-016-3891-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 01/15/2016] [Indexed: 01/09/2023]
Abstract
Mitochondria fulfil multiple tasks in nutrient metabolism, energy production, redox homeostasis and stress response, and are essential for pancreatic beta cell function. The dynamism and health of the mitochondrial network is regulated by fission- and fusion-triggering factors and by a quality control system that removes dysfunctional organelles. Alongside the role of mitochondria in regulating apoptotic cell death mediated primarily via production of reactive oxygen species and release of cytochrome c, there is evidence of other links between mitochondria and inflammation that have implications for cell viability. This review briefly outlines two pathways that are potentially vital for pancreatic beta cell function. The first concerns the regulation of Parkin, a protein that acts, not only as a central player in regulating mitophagy, but also as an activator of the NF-ĸB pathway. The fact that expression of optic atrophy protein 1 (OPA1), a mitochondrial fusion inducer and master mitochondrial cristae biogenetic factor, is increased following NF-ĸB activation highlights a point of mitochondrial control that might be influenced by TNFα signalling. A second axis of interest is suggested by IL-6-mediated upregulation of the fission inducer FIS1 alongside downregulation of mitofusin 2 (MFN2), a guard of mitochondrial fusion and metabolism and an inhibitor of apoptosis. This review summarises a presentation given at the 'Islet inflammation in type 2 diabetes' symposium at the 2015 annual meeting of the EASD. It is accompanied two other reviews on topics from this symposium (by Marc Donath, DOI: 10.1007/s00125-016-3873-z , and Jerry Nadler and colleagues, DOI: 10.1007/s00125-016-3890-y ) and a commentary by the Session Chair, Piero Marchetti (DOI: 10.1007/s00125-016-3875-x ).
Collapse
Affiliation(s)
- Simone Baltrusch
- Institute of Medical Biochemistry and Molecular Biology, University of Rostock, Schillingallee 70, 18057, Rostock, Germany.
| |
Collapse
|
28
|
Nguyen N, Bellile E, Thomas D, McHugh J, Rozek L, Virani S, Peterson L, Carey TE, Walline H, Moyer J, Spector M, Perim D, Prince M, McLean S, Bradford CR, Taylor JMG, Wolf GT. Tumor infiltrating lymphocytes and survival in patients with head and neck squamous cell carcinoma. Head Neck 2016; 38:1074-84. [PMID: 26879675 DOI: 10.1002/hed.24406] [Citation(s) in RCA: 259] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2015] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Because immune responses within the tumor microenvironment are important predictors of tumor biology, correlations of types of tumor infiltrating lymphocytes (TILs) with clinical outcomes were determined in 278 patients with head and neck squamous cell carcinoma (HNSCC). METHODS Infiltrating levels of CD4 (helper T cells), CD8 (cytotoxic/suppressor T cells), FoxP3 (regulatory T cells), CD68 (myeloid-derived suppressor cells,) and CD1a (Langerhans) cells were measured in tissue microarrays (TMAs). Cox models tested associations with patient outcomes after adjusting for all known prognostic factors. Median follow-up was 36.6 months. RESULTS Higher CD4 and CD8 TIL levels were associated with improved overall survival (OS; hazard ratio [HR] = 0.77; 95% confidence interval [CI] = 0.65-0.93; p = .005 and HR = 0.77; 95% CI = 0.64-0.94; p = .008, respectively), and relapse-free survival (RFS; p = .03 and .05, respectively). After controlling for prognostic factors, higher CD4 levels predicted improved OS and disease-specific survival (DSS; p = .003 and p = .004, respectively). CONCLUSION The findings suggest that TILs are a significant independent prognostic factor for HNSCC that differ by treatment. © 2016 Wiley Periodicals, Inc. Head Neck 38: 1074-1084, 2016.
Collapse
Affiliation(s)
- Nghia Nguyen
- Department of Otolaryngology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Emily Bellile
- Center for Cancer Biostatistics, Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Daffyd Thomas
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jonathan McHugh
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Laura Rozek
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Shama Virani
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Lisa Peterson
- Department of Otolaryngology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Thomas E Carey
- Department of Otolaryngology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Heather Walline
- Department of Otolaryngology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jeffery Moyer
- Department of Otolaryngology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Matthew Spector
- Department of Otolaryngology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Daniel Perim
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, Michigan
| | - Mark Prince
- Department of Otolaryngology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Scott McLean
- Department of Otolaryngology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Carol R Bradford
- Department of Otolaryngology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jeremy M G Taylor
- Center for Cancer Biostatistics, Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Gregory T Wolf
- Department of Otolaryngology, University of Michigan Medical School, Ann Arbor, Michigan
| | | |
Collapse
|
29
|
Rütti S, Howald C, Arous C, Dermitzakis E, Halban PA, Bouzakri K. IL-13 improves beta-cell survival and protects against IL-1beta-induced beta-cell death. Mol Metab 2015; 5:122-131. [PMID: 26909320 PMCID: PMC4735661 DOI: 10.1016/j.molmet.2015.11.003] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 11/04/2015] [Accepted: 11/09/2015] [Indexed: 12/25/2022] Open
Abstract
Objectives IL-13 is a cytokine classically produced by anti-inflammatory T-helper-2 lymphocytes; it is decreased in the circulation of type 2 diabetic patients and impacts positively on liver and skeletal muscle. Although IL-13 can exert positive effects on beta-cell lines, its impact and mode of action on primary beta-cell function and survival remain largely unexplored. Methods Beta-cells were cultured for 48 h in the presence of IL-13 alone or in combination with IL-1β or cytokine cocktail (IL-1β, IFNγ, TNFα). Results IL-13 protected human and rat beta-cells against cytokine induced death. However, IL-13 was unable to protect from IL-1β impaired glucose stimulated insulin secretion and did not influence NFκB nuclear relocalization induced by IL-1β. IL-13 induced phosphorylation of Akt, increased IRS2 protein expression and counteracted the IL-1β induced regulation of several beta-cell stress response genes. Conclusions The prosurvival effects of IL-13 thus appear to be mediated through IRS2/Akt signaling with NFκB independent regulation of gene expression. In addition to previously documented beneficial effects on insulin target tissues, these data suggest that IL-13 may be useful for treatment of type 2 diabetes by preserving beta-cell mass or slowing its rate of decline. IL-13 decreases human beta-cells apoptosis. IL-13 protects primary beta-cells form cytokine induced apoptosis. The IRS2/akt pathway mediates IL-13 protective effects. IL-13 modulates the expression of genes involved in stress response.
Collapse
Affiliation(s)
- Sabine Rütti
- Department of Genetic Medicine and Development, University of Geneva Medical Center, Geneva, Switzerland.
| | - Cédric Howald
- Department of Genetic Medicine and Development, University of Geneva Medical Center, Geneva, Switzerland
| | - Caroline Arous
- Department of Genetic Medicine and Development, University of Geneva Medical Center, Geneva, Switzerland
| | - Emmanouil Dermitzakis
- Department of Genetic Medicine and Development, University of Geneva Medical Center, Geneva, Switzerland
| | - Philippe A Halban
- Department of Genetic Medicine and Development, University of Geneva Medical Center, Geneva, Switzerland
| | - Karim Bouzakri
- Department of Genetic Medicine and Development, University of Geneva Medical Center, Geneva, Switzerland
| |
Collapse
|
30
|
Kiba T. Ventromedial hypothalamic lesions downregulate multiple immune signaling pathways in rat pancreatic islets. Neurosci Lett 2015; 610:177-81. [PMID: 26555798 DOI: 10.1016/j.neulet.2015.11.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 11/04/2015] [Accepted: 11/04/2015] [Indexed: 12/17/2022]
Abstract
It was recently reported that ventromedial hypothalamic lesions change the expression of cell proliferation-related genes and metabolism-related genes in rat pancreatic islets. This study has examined how gene families involved in immune responses are regulated in rat pancreatic islets after VMH lesions formation. Total pancreatic islets RNA was extracted, and differences in the gene expression profiles between rats at day 3 after VMH lesioning and sham-VMH-lesioned rats were investigated using DNA microarray and real-time polymerase chain reaction. VMH lesions downregulated multiple immune signaling pathways in rat pancreatic islets. Real-time polymerase chain reaction also confirmed that gene expressions of RT1 class II, locus Bb (RT1-Bb) was up-regulated and Spi-B transcription factor (Spib) was downregulated at day 3 after the VMH lesions. Ventromedial hypothalamic lesions may change the expression of multiple immune response genes in rat pancreatic islets.
Collapse
Affiliation(s)
- Takayoshi Kiba
- Division of Modern Medical Technology, Institute for Clinical Research, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Kure 737-0023, Japan.
| |
Collapse
|
31
|
Christensen CS, Christensen DP, Lundh M, Dahllöf MS, Haase TN, Velasquez JM, Laye MJ, Mandrup-Poulsen T, Solomon TPJ. Skeletal Muscle to Pancreatic β-Cell Cross-talk: The Effect of Humoral Mediators Liberated by Muscle Contraction and Acute Exercise on β-Cell Apoptosis. J Clin Endocrinol Metab 2015. [PMID: 26218753 DOI: 10.1210/jc.2014-4506] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
CONTEXT Mechanisms explaining exercise-induced β-cell health are unknown. OBJECTIVE This study aimed to define the role of muscle contraction and acute exercise-derived soluble humoral mediators on β-cell health. DESIGN In vitro models were used. SETTING University. PARTICIPANTS Healthy subjects. INTERVENTION(S) Conditioned media (CM) were collected from human skeletal muscle (HSkM) cells treated with or without electrical pulse stimulation (EPS). Antecubital and femoral venous blood serum were collected before and after an exercise bout. CM and sera with or without IL-6 neutralization were used to incubate insulin-producing INS-1 cells and rat islets for 24 h in the presence or absence of proinflammatory cytokines (IL-1β+IFN-γ). MAIN OUTCOME MEASURE(S) INS-1 and islet apoptosis and accumulated insulin secretion. RESULTS IL-1β+IFN-γ increased INS-1 and islet apoptosis and decreased insulin secretion. EPS-treated HSkM cell CM did not affect these variables. Exercise-conditioned antecubital but not femoral sera prevented IL-1β+IFN-γ-induced INS-1 and islet apoptosis. Femoral sera reduced insulin secretion under normal and proinflammatory conditions in INS-1 but not islet cells. EPS increased HSkM cell IL-6 secretion and exercise increased circulating IL-6 levels in antecubital and femoral serum. IL-6 neutralization demonstrated that muscle-derived IL-6 prevents INS-1 and islet apoptosis in the absence of IL-1β+IFN-γ, but augments apoptosis under proinflammatory conditions, and that muscle-derived IL-6 supports islet insulin secretion in the absence of IL-1β+IFN-γ. CONCLUSIONS Unidentified circulating humoral mediators released during exercise prevent proinflammatory cytokine-induced β-cell apoptosis. Muscle-derived mediators released during exercise suppress β-cell insulin secretion. Furthermore, muscle-derived IL-6 seems to prevent β-cell apoptosis under normal conditions but contributes to β-cell apoptosis under proinflammatory conditions.
Collapse
Affiliation(s)
- Camilla S Christensen
- Department of Biomedical Sciences (C.S.C., D.P.C., M.S.D., T.N.H., J.M.V., T.M.-P.), Panum Institute, University of Copenhagen, 2200, Denmark; Centre for Inflammation and Metabolism (J.M.V., M.J.L.), Rigshospitalet, Copenhagen, 2100, Denmark; Department of Molecular Medicine and Surgery (J.M.-P.), Karolinska Institutet, Stockholm, SE-171 77, Sweden; and School of Sport, Exercise, and Rehabilitation Sciences (T.P.J.S.), Centre for Endocrinology, Diabetes, and Metabolism, University of Birmingham, B15 2TT, United Kingdom
| | - Dan P Christensen
- Department of Biomedical Sciences (C.S.C., D.P.C., M.S.D., T.N.H., J.M.V., T.M.-P.), Panum Institute, University of Copenhagen, 2200, Denmark; Centre for Inflammation and Metabolism (J.M.V., M.J.L.), Rigshospitalet, Copenhagen, 2100, Denmark; Department of Molecular Medicine and Surgery (J.M.-P.), Karolinska Institutet, Stockholm, SE-171 77, Sweden; and School of Sport, Exercise, and Rehabilitation Sciences (T.P.J.S.), Centre for Endocrinology, Diabetes, and Metabolism, University of Birmingham, B15 2TT, United Kingdom
| | - Morten Lundh
- Department of Biomedical Sciences (C.S.C., D.P.C., M.S.D., T.N.H., J.M.V., T.M.-P.), Panum Institute, University of Copenhagen, 2200, Denmark; Centre for Inflammation and Metabolism (J.M.V., M.J.L.), Rigshospitalet, Copenhagen, 2100, Denmark; Department of Molecular Medicine and Surgery (J.M.-P.), Karolinska Institutet, Stockholm, SE-171 77, Sweden; and School of Sport, Exercise, and Rehabilitation Sciences (T.P.J.S.), Centre for Endocrinology, Diabetes, and Metabolism, University of Birmingham, B15 2TT, United Kingdom
| | - Mattias S Dahllöf
- Department of Biomedical Sciences (C.S.C., D.P.C., M.S.D., T.N.H., J.M.V., T.M.-P.), Panum Institute, University of Copenhagen, 2200, Denmark; Centre for Inflammation and Metabolism (J.M.V., M.J.L.), Rigshospitalet, Copenhagen, 2100, Denmark; Department of Molecular Medicine and Surgery (J.M.-P.), Karolinska Institutet, Stockholm, SE-171 77, Sweden; and School of Sport, Exercise, and Rehabilitation Sciences (T.P.J.S.), Centre for Endocrinology, Diabetes, and Metabolism, University of Birmingham, B15 2TT, United Kingdom
| | - Tobias N Haase
- Department of Biomedical Sciences (C.S.C., D.P.C., M.S.D., T.N.H., J.M.V., T.M.-P.), Panum Institute, University of Copenhagen, 2200, Denmark; Centre for Inflammation and Metabolism (J.M.V., M.J.L.), Rigshospitalet, Copenhagen, 2100, Denmark; Department of Molecular Medicine and Surgery (J.M.-P.), Karolinska Institutet, Stockholm, SE-171 77, Sweden; and School of Sport, Exercise, and Rehabilitation Sciences (T.P.J.S.), Centre for Endocrinology, Diabetes, and Metabolism, University of Birmingham, B15 2TT, United Kingdom
| | - Jessica M Velasquez
- Department of Biomedical Sciences (C.S.C., D.P.C., M.S.D., T.N.H., J.M.V., T.M.-P.), Panum Institute, University of Copenhagen, 2200, Denmark; Centre for Inflammation and Metabolism (J.M.V., M.J.L.), Rigshospitalet, Copenhagen, 2100, Denmark; Department of Molecular Medicine and Surgery (J.M.-P.), Karolinska Institutet, Stockholm, SE-171 77, Sweden; and School of Sport, Exercise, and Rehabilitation Sciences (T.P.J.S.), Centre for Endocrinology, Diabetes, and Metabolism, University of Birmingham, B15 2TT, United Kingdom
| | - Matthew J Laye
- Department of Biomedical Sciences (C.S.C., D.P.C., M.S.D., T.N.H., J.M.V., T.M.-P.), Panum Institute, University of Copenhagen, 2200, Denmark; Centre for Inflammation and Metabolism (J.M.V., M.J.L.), Rigshospitalet, Copenhagen, 2100, Denmark; Department of Molecular Medicine and Surgery (J.M.-P.), Karolinska Institutet, Stockholm, SE-171 77, Sweden; and School of Sport, Exercise, and Rehabilitation Sciences (T.P.J.S.), Centre for Endocrinology, Diabetes, and Metabolism, University of Birmingham, B15 2TT, United Kingdom
| | - Thomas Mandrup-Poulsen
- Department of Biomedical Sciences (C.S.C., D.P.C., M.S.D., T.N.H., J.M.V., T.M.-P.), Panum Institute, University of Copenhagen, 2200, Denmark; Centre for Inflammation and Metabolism (J.M.V., M.J.L.), Rigshospitalet, Copenhagen, 2100, Denmark; Department of Molecular Medicine and Surgery (J.M.-P.), Karolinska Institutet, Stockholm, SE-171 77, Sweden; and School of Sport, Exercise, and Rehabilitation Sciences (T.P.J.S.), Centre for Endocrinology, Diabetes, and Metabolism, University of Birmingham, B15 2TT, United Kingdom
| | - Thomas P J Solomon
- Department of Biomedical Sciences (C.S.C., D.P.C., M.S.D., T.N.H., J.M.V., T.M.-P.), Panum Institute, University of Copenhagen, 2200, Denmark; Centre for Inflammation and Metabolism (J.M.V., M.J.L.), Rigshospitalet, Copenhagen, 2100, Denmark; Department of Molecular Medicine and Surgery (J.M.-P.), Karolinska Institutet, Stockholm, SE-171 77, Sweden; and School of Sport, Exercise, and Rehabilitation Sciences (T.P.J.S.), Centre for Endocrinology, Diabetes, and Metabolism, University of Birmingham, B15 2TT, United Kingdom
| |
Collapse
|
32
|
An Autocrine Cytokine/JAK/STAT-Signaling Induces Kynurenine Synthesis in Multidrug Resistant Human Cancer Cells. PLoS One 2015; 10:e0126159. [PMID: 25955018 PMCID: PMC4425697 DOI: 10.1371/journal.pone.0126159] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 03/29/2015] [Indexed: 12/01/2022] Open
Abstract
Background Multidrug resistant cancer cells are hard to eradicate for the inefficacy of conventional anticancer drugs. Besides escaping the cytotoxic effects of chemotherapy, they also bypass the pro-immunogenic effects induced by anticancer drugs: indeed they are not well recognized by host dendritic cells and do not elicit a durable anti-tumor immunity. It has not yet been investigated whether multidrug resistant cells have a different ability to induce immunosuppression than chemosensitive ones. We addressed this issue in human and murine chemosensitive and multidrug resistant cancer cells. Results We found that the activity and expression of indoleamine 2,3-dioxygenase 1 (IDO1), which catalyzes the conversion of tryptophan into the immunosuppressive metabolite kynurenine, was higher in all the multidrug resistant cells analyzed and that IDO1 inhibition reduced the growth of drug-resistant tumors in immunocompetent animals. In chemoresistant cells the basal activity of JAK1/STAT1 and JAK1/STAT3 signaling was higher, the STAT3 inhibitor PIAS3 was down-regulated, and the autocrine production of STAT3-target and IDO1-inducers cytokines IL-6, IL-4, IL-1β, IL-13, TNF-α and CD40L, was increased. The disruption of the JAK/STAT signaling lowered the IDO1 activity and reversed the kynurenine-induced pro-immunosuppressive effects, as revealed by the restored proliferation of T-lymphocytes in STAT-silenced chemoresistant cells. Conclusions Our work shows that multidrug resistant cells have a stronger immunosuppressive attitude than chemosensitive cells, due to the constitutive activation of the JAK/STAT/IDO1 axis, thus resulting chemo- and immune-evasive. Disrupting this axis may significantly improve the efficacy of chemo-immunotherapy protocols against resistant tumors.
Collapse
|
33
|
Padgett LE, Burg AR, Lei W, Tse HM. Loss of NADPH oxidase-derived superoxide skews macrophage phenotypes to delay type 1 diabetes. Diabetes 2015; 64:937-46. [PMID: 25288672 PMCID: PMC4338593 DOI: 10.2337/db14-0929] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Macrophages are early islet-infiltrating cells seen in type 1 diabetes (T1D). While proinflammatory M1 macrophages induce T1D, M2 macrophages have been shown to delay this autoimmune disease in nonobese diabetic (NOD) mice, but the environmental cues that govern macrophage polarization and differentiation remain unresolved. We previously demonstrated the importance of reactive oxygen species (ROS) in T1D, as NOD mice deficient in NADPH oxidase (NOX)-derived superoxide (Ncf1(m1J)) were protected against T1D partly because of blunted Toll-like receptor-dependent macrophage responses. We provide evidence that NOX-derived ROS contribute to macrophage differentiation in T1D. During spontaneous diabetes progression, T1D-resistant NOD.Ncf1(m1J) islet-resident macrophages displayed a dampened M1 and increased M2 phenotype. The transfer of diabetogenic T cells into NOX-deficient NOD.Rag.Ncf1(m1J) recipients resulted in decreased TNF-α(+) and IL-1β(+) islet-infiltrating M1 macrophages and a concomitant enhancement in arginase-1(+) M2 macrophages. Mechanistic analysis of superoxide-deficient bone marrow-derived macrophages revealed a marked diminution in a proinflammatory M1 phenotype due to decreased P-STAT1 (Y701) and interferon regulatory factor 5 compared with NOD mice. We have therefore defined a novel mechanistic link between NOX-derived ROS and macrophage phenotypes, and implicated superoxide as an important factor in macrophage differentiation. Thus, targeting macrophage redox status may represent a promising therapy in halting human T1D.
Collapse
Affiliation(s)
- Lindsey E Padgett
- Department of Microbiology, Comprehensive Diabetes Center, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Ashley R Burg
- Department of Microbiology, Comprehensive Diabetes Center, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Weiqi Lei
- Department of Microbiology, Comprehensive Diabetes Center, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Hubert M Tse
- Department of Microbiology, Comprehensive Diabetes Center, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
34
|
Melnik BC. The pathogenic role of persistent milk signaling in mTORC1- and milk-microRNA-driven type 2 diabetes mellitus. Curr Diabetes Rev 2015; 11:46-62. [PMID: 25587719 PMCID: PMC4428476 DOI: 10.2174/1573399811666150114100653] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 01/08/2015] [Accepted: 01/09/2015] [Indexed: 12/12/2022]
Abstract
Milk, the secretory product of the lactation genome, promotes growth of the newborn mammal. Milk delivers insulinotropic amino acids, thus maintains a molecular crosstalk with the pancreatic β-cell of the milk recipient. Homeostasis of β-cells and insulin production depend on the appropriate magnitude of mTORC1 signaling. mTORC1 is activated by branched-chain amino acids (BCAAs), glutamine, and palmitic acid, abundant nutrient signals of cow´s milk. Furthermore, milk delivers bioactive exosomal microRNAs. After milk consumption, bovine microRNA-29b, a member of the diabetogenic microRNA-29- family, reaches the systemic circulation and the cells of the milk consumer. MicroRNA-29b downregulates branchedchain α-ketoacid dehydrogenase, a potential explanation for increased BCAA serum levels, the metabolic signature of insulin resistance and type 2 diabetes mellitus (T2DM). In non-obese diabetic mice, microRNA-29b downregulates the antiapoptotic protein Mcl-1, which leads to early β-cell death. In all mammals except Neolithic humans, milk-driven mTORC1 signaling is physiologically restricted to the postnatal period. In contrast, chronic hyperactivated mTORC1 signaling has been associated with the development of age-related diseases of civilization including T2DM. Notably, chronic hyperactivation of mTORC1 enhances endoplasmic reticulum stress that promotes apoptosis. In fact, hyperactivated β-cell mTORC1 signaling induced early β-cell apoptosis in a mouse model. The EPIC-InterAct Study demonstrated an association between milk consumption and T2DM in France, Italy, United Kingdom, Germany, and Sweden. In contrast, fermented milk products and cheese exhibit an inverse correlation. Since the early 1950´s, refrigeration technology allowed widespread consumption of fresh pasteurized milk, which facilitates daily intake of bioactive bovine microRNAs. Persistent uptake of cow´s milk-derived microRNAs apparently transfers an overlooked epigenetic diabetogenic program that should not reach the human food chain.
Collapse
Affiliation(s)
- Bodo C Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabruck, Sedanstraße 115, D-49090 Osnabrück, Germany.
| |
Collapse
|
35
|
Flanagan SE, Haapaniemi E, Russell MA, Caswell R, Allen HL, De Franco E, McDonald TJ, Rajala H, Ramelius A, Barton J, Heiskanen K, Heiskanen-Kosma T, Kajosaari M, Murphy NP, Milenkovic T, Seppänen M, Lernmark Å, Mustjoki S, Otonkoski T, Kere J, Morgan NG, Ellard S, Hattersley AT. Activating germline mutations in STAT3 cause early-onset multi-organ autoimmune disease. Nat Genet 2014; 46:812-814. [PMID: 25038750 PMCID: PMC4129488 DOI: 10.1038/ng.3040] [Citation(s) in RCA: 379] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 06/27/2014] [Indexed: 12/15/2022]
Abstract
Monogenic causes of autoimmunity provide key insights into the complex regulation of the immune system. We report a new monogenic cause of autoimmunity resulting from de novo germline activating STAT3 mutations in five individuals with a spectrum of early-onset autoimmune disease, including type 1 diabetes. These findings emphasize the critical role of STAT3 in autoimmune disease and contrast with the germline inactivating STAT3 mutations that result in hyper IgE syndrome.
Collapse
Affiliation(s)
- Sarah E. Flanagan
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, EX2 5DW, UK
| | - Emma Haapaniemi
- Folkhälsan Institute of Genetics, University of Helsinki, Helsinki, Finland
- Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland
| | - Mark A. Russell
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, EX2 5DW, UK
| | - Richard Caswell
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, EX2 5DW, UK
| | - Hana Lango Allen
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, EX2 5DW, UK
| | - Elisa De Franco
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, EX2 5DW, UK
| | - Timothy J. McDonald
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, EX2 5DW, UK
| | - Hanna Rajala
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki
- Helsinki University Central Hospital Cancer Center, Helsinki, Finland
| | - Anita Ramelius
- Department of Clinical Sciences, Lund University, Lund, Sweden
- CRC, Skåne University Hospital SUS, Malmö, Sweden
| | - John Barton
- Bristol Royal Hospital for Children, Upper Maudlin Street, Bristol, BS2 8BJ, UK
| | - Kaarina Heiskanen
- Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland
- Children’s Hospital, Helsinki University Central Hospital, Helsinki, Finland
| | | | - Merja Kajosaari
- Children’s Hospital, Helsinki University Central Hospital, Helsinki, Finland
| | - Nuala P. Murphy
- Department of Diabetes and Endocrinology, Children’s University Hospital, Temple St., Dublin 1, Ireland
| | - Tatjana Milenkovic
- Department of Endocrinology, Institute for Mother and Child Health Care of Serbia ‘Dr Vukan Cupic’, Belgrade, Serbia
| | - Mikko Seppänen
- Immunodeficiency Unit, Division of Infectious Diseases, Helsinki University Central Hospital, Helsinki, Finland
| | - Åke Lernmark
- Department of Clinical Sciences, Lund University, Lund, Sweden
- CRC, Skåne University Hospital SUS, Malmö, Sweden
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki
- Helsinki University Central Hospital Cancer Center, Helsinki, Finland
| | - Timo Otonkoski
- Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland
- Children’s Hospital, Helsinki University Central Hospital, Helsinki, Finland
| | - Juha Kere
- Folkhälsan Institute of Genetics, University of Helsinki, Helsinki, Finland
- Research Programs Unit, Molecular Neurology, University of Helsinki, Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Hälsovägen 7, 14183 Huddinge, Sweden
- Center for Innovative Medicine, Karolinska Institutet, Hälsovägen 7, 14183 Huddinge, Sweden
| | - Noel G. Morgan
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, EX2 5DW, UK
| | - Sian Ellard
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, EX2 5DW, UK
| | - Andrew T. Hattersley
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, EX2 5DW, UK
| |
Collapse
|
36
|
Arzt L, Kothmaier H, Halbwedl I, Quehenberger F, Popper HH. Signal transducer and activator of transcription 1 (STAT1) acts like an oncogene in malignant pleural mesothelioma. Virchows Arch 2014; 465:79-88. [DOI: 10.1007/s00428-014-1584-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 03/17/2014] [Accepted: 04/22/2014] [Indexed: 11/28/2022]
|
37
|
Abstract
Considerable efforts have been invested to understand the mechanisms by which pro-inflammatory cytokines mediate the demise of β-cells in type 1 diabetes but much less attention has been paid to the role of anti-inflammatory cytokines as potential cytoprotective agents in these cells. Despite this, there is increasing evidence that anti-inflammatory molecules such as interleukin (IL)-4, IL-10 and IL-13 can exert a direct influence of β-cell function and viability and that the circulating levels of these cytokines may be reduced in type 1 diabetes. Thus, it seems possible that targeting of anti-inflammatory pathways might offer therapeutic potential in this disease. In the present review, we consider the evidence implicating IL-4, IL-10 and IL-13 as cytoprotective agents in the β-cell and discuss the receptor components and downstream signaling pathways that mediate these effects.
Collapse
Affiliation(s)
- M A Russell
- Institute of Biomedical and Clinical Science; University of
Exeter Medical School; Exeter, Devon, UK
- Correspondence to: MA
Russell;
| | - N G Morgan
- Institute of Biomedical and Clinical Science; University of
Exeter Medical School; Exeter, Devon, UK
| |
Collapse
|
38
|
Wang Y. Attenuation of berberine on lipopolysaccharide-induced inflammatory and apoptosis responses in β-cells via TLR4-independent JNK/NF-κB pathway. PHARMACEUTICAL BIOLOGY 2013; 52:532-538. [PMID: 24188583 DOI: 10.3109/13880209.2013.840851] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Abstract Context: Toll-like receptor 4 (TLR4)-independent inflammatory and apoptosis responses contribute to β-cell failure in diabetes mellitus (DM). Berberine (BBR), a bioactive isoquinoline derivative alkaloid, ameliorates the inflammatory response in DM. Objective: This study explored the protective mechanisms of BBR on TLR4-independent inflammation response in β cells. Materials and methods: Lipopolysaccharide (LPS; 100 ng/ml) was used to induce the inflammatory response in NIT-1 and rat insulinoma (INS-1) cells for 24 h. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and colony formation assays were used for the determination of cell viability. The levels of monocyte chemoattractant protein (MCP-1), interleukin 6 (IL-6), tumor necrosis factor α (TNF-α) and insulin in cultured supernatant were detected by enzyme-linked immunosorbent assay kits. Western blot analysis was performed for the expression of p-c-Jun N-terminal kinase (JNK) and p65 NF-κB in NIT-1 cells, and p65 NF-κB in INS-1 cells. Results: BBR (1.25, 2.5 and 5 μM) or TLR4 inhibitor (TAK-242, 1 μM) increased remarkably NIT-1 cell viability by 72.6 ± 5.0, 85.9 ± 9.3, 94.7 ± 7.1 and 92.6 ± 8.4%. The EC50 of BBR was 1.14 μM. Colony formation assay showed that BBR increased the number of colonies of NIT-1 and INS-1 cells. BBR, TAK-242 or SP-600125 (1 μM) could significantly reduce the levels of MCP-1, IL-6 and TNF-α, insulin and JNK and NF-κB phosphorylation in NIT-1 cells, as well as the p65 NF-κB in INS-1 cells. Discussion and conclusion: BBR could ameliorate LPS-induced β-cell injury through the TLR4-independent JNK/NF-κB pathway. Thus, this pathway may be a potential target for the prevention and treatment of DM.
Collapse
Affiliation(s)
- Ying Wang
- Department of Endocrinology, Tianjin Pingjin Hospital , Tianjin , P.R. China
| |
Collapse
|
39
|
Solomon TPJ, Malin SK, Karstoft K, Kashyap SR, Haus JM, Kirwan JP. Pancreatic β-cell function is a stronger predictor of changes in glycemic control after an aerobic exercise intervention than insulin sensitivity. J Clin Endocrinol Metab 2013; 98:4176-86. [PMID: 23966244 PMCID: PMC3790622 DOI: 10.1210/jc.2013-2232] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
CONTEXT Understanding intersubject variability in glycemic control following exercise training will help individualize treatment. OBJECTIVE Our aim was to determine whether this variability is related to training-induced changes in insulin sensitivity or pancreatic β-cell function. DESIGN, SETTING, AND PARTICIPANTS We conducted an observational clinical study of 105 subjects with impaired glucose tolerance or type 2 diabetes. INTERVENTIONS AND MAIN OUTCOME MEASURES Individual subject changes in fitness (VO2max), glycemia (glycosylated hemoglobin, fasting glucose, oral glucose tolerance test), insulin sensitivity (hyperinsulinemic-euglycemic clamp), oral glucose-stimulated insulin secretion (GSIS), and disposition index (DI) were measured following 12 to 16 weeks of aerobic exercise training. Regression analyses were used to identify relationships between variables. RESULTS After training, 86% of subjects increased VO2max and lost weight. Glycosylated hemoglobin, fasting glucose, and 2-hour oral glucose tolerance test were reduced in 69%, 62%, and 68% of subjects, respectively, while insulin sensitivity improved in 90% of the participants. Changes in glycemic control were congruent with changes in GSIS such that 66% of subjects had a reduction in first-phase GSIS, and 46% had reduced second-phase GSIS. Training increased first- and second-phase DI in 83% and 74% of subjects. Training-induced changes in glycemic control were related to changes in GSIS (P < .05), but not insulin sensitivity or DI, and training-induced improvements in glycemic control were largest in subjects with greater pretraining GSIS. CONCLUSIONS Intersubject variability in restoring glycemic control following exercise is explained primarily by changes in insulin secretion. Thus, baseline and training-induced changes in β-cell function may be a key determinant of training-induced improvements in glycemic control.
Collapse
Affiliation(s)
- Thomas P J Solomon
- PhD, Department of Biomedical Sciences, Panum Institute 4.5.13, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark.
| | | | | | | | | | | |
Collapse
|