1
|
Aglan HA, Kotob SE, Mahmoud NS, Kishta MS, Ahmed HH. Bone marrow stem cell-derived β-cells: New issue for diabetes cell therapy. Tissue Cell 2024; 86:102280. [PMID: 38029457 DOI: 10.1016/j.tice.2023.102280] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/19/2023] [Accepted: 11/20/2023] [Indexed: 12/01/2023]
Abstract
This investigation aimed to establish the promising role of insulin-producing cells (IPCs) growing from bone marrow-mesenchymal stem cells (BM-MSCs) in relieving hyperglycemia induced in rats. BM-MSCs were differentiated into IPCs using three different protocols. The efficiency of BM-MSCs differentiation into IPCs in vitro was confirmed by detecting IPCs specific gene expression (Foxa-2, PDX-1 and Ngn-3) and insulin release assay. The in vivo study design included 3 groups of male Wistar rats; negative control group, diabetic group and IPCs-transfused group (5 ×106 cells of the most functional IPCs/rat). One month after IPCs infusion, serum glucose, insulin, c-peptide and visfatin levels as well as pancreatic glucagon level were quantified. Gene expression analysis of pancreatic Foxa-2 and Sox-17, IGF-1 and FGF-10 was done. Additionally, histological investigation of pancreatic tissue sections was performed. Our data clarified that, the most functional IPCs are those generated from BM-MSCs using differentiation protocol 3 as indicated by the significant up-regulation of Foxa-2, PDX-1 and Ngn-3 gene expression levels. These findings were further emphasized by releasing of a significant amount of insulin in response to glucose load. The transplantation of the IPCs in diabetic rats elicited significant decline in serum glucose, visfatin and pancreatic glucagon levels along with significant rise in serum insulin and c-peptide levels. Moreover, it triggered significant up-regulation in the expression levels of pancreatic Foxa-2, Sox-17, IGF-1 and FGF-10 genes versus the untreated diabetic counterpart. The histopathological examination of pancreatic tissue almost assisted the biochemical and molecular genetic analyses. These results disclose that the cell therapy holds potential to develop a new cure for DM based on the capability of BM-MSCs to generate β-cell phenotype using specific protocol.
Collapse
Affiliation(s)
- Hadeer A Aglan
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt; Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Giza, Egypt.
| | - Soheir E Kotob
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt
| | - Nadia S Mahmoud
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt; Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Giza, Egypt
| | - Mohamed S Kishta
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt; Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Giza, Egypt
| | - Hanaa H Ahmed
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt; Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Giza, Egypt
| |
Collapse
|
2
|
Kaminitz A, Ash S, Askenasy N. Neutralization Versus Reinforcement of Proinflammatory Cytokines to Arrest Autoimmunity in Type 1 Diabetes. Clin Rev Allergy Immunol 2018; 52:460-472. [PMID: 27677500 DOI: 10.1007/s12016-016-8587-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
As physiological pathways of intercellular communication produced by all cells, cytokines are involved in the pathogenesis of inflammatory insulitis as well as pivotal mediators of immune homeostasis. Proinflammatory cytokines including interleukins, interferons, transforming growth factor-β, tumor necrosis factor-α, and nitric oxide promote destructive insulitis in type 1 diabetes through amplification of the autoimmune reaction, direct toxicity to β-cells, and sensitization of islets to apoptosis. The concept that neutralization of cytokines may be of therapeutic benefit has been tested in few clinical studies, which fell short of inducing sustained remission or achieving disease arrest. Therapeutic failure is explained by the redundant activities of individual cytokines and their combinations, which are rather dispensable in the process of destructive insulitis because other cytolytic pathways efficiently compensate their deficiency. Proinflammatory cytokines are less redundant in regulation of the inflammatory reaction, displaying protective effects through restriction of effector cell activity, reinforcement of suppressor cell function, and participation in islet recovery from injury. Our analysis suggests that the role of cytokines in immune homeostasis overrides their contribution to β-cell death and may be used as potent immunomodulatory agents for therapeutic purposes rather than neutralized.
Collapse
Affiliation(s)
- Ayelet Kaminitz
- The Leah and Edward M. Frankel Laboratory of Experimental Bone Marrow Transplantation, 14 Kaplan Street, Petach Tikva, Israel, 49202
| | - Shifra Ash
- The Leah and Edward M. Frankel Laboratory of Experimental Bone Marrow Transplantation, 14 Kaplan Street, Petach Tikva, Israel, 49202
| | - Nadir Askenasy
- The Leah and Edward M. Frankel Laboratory of Experimental Bone Marrow Transplantation, 14 Kaplan Street, Petach Tikva, Israel, 49202.
| |
Collapse
|
3
|
Yolcu ES, Shirwan H, Askenasy N. Fas/Fas-Ligand Interaction As a Mechanism of Immune Homeostasis and β-Cell Cytotoxicity: Enforcement Rather Than Neutralization for Treatment of Type 1 Diabetes. Front Immunol 2017; 8:342. [PMID: 28396667 PMCID: PMC5366321 DOI: 10.3389/fimmu.2017.00342] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 03/09/2017] [Indexed: 12/22/2022] Open
Affiliation(s)
- Esma S Yolcu
- Department of Microbiology and Immunology, Institute for Cellular Therapeutics, University of Louisville , Louisville, KY , USA
| | - Haval Shirwan
- Department of Microbiology and Immunology, Institute for Cellular Therapeutics, University of Louisville , Louisville, KY , USA
| | - Nadir Askenasy
- Frankel Laboratory of Experimental Bone Marrow Transplantation , Petach Tikva , Israel
| |
Collapse
|
4
|
Abstract
OBJECTIVES This research study sought to improve the treatment of pancreatic cancer by improving the drug delivery of a promising AKT/PDK1 inhibitor, PHT-427, in poly(lactic-co-glycolic) acid (PLGA) nanoparticles. METHODS PHT-427 was encapsulated in single-emulsion and double-emulsion PLGA nanoparticles (SE-PLGA-427 and DE-PLGA-427). The drug release rate was evaluated to assess the effect of the second PLGA layer of DE-PLGA-427. Ex vivo cryo-imaging and drug extraction from ex vivo organs was used to assess the whole-body biodistribution in an orthotopic model of MIA PaCa-2 pancreatic cancer. Anatomical magnetic resonance imaging (MRI) was used to noninvasively assess the effects of 4 weeks of nanoparticle drug treatment on tumor size, and diffusion-weighted MRI longitudinally assessed changes in tumor cellularity. RESULTS DE-PLGA-427 showed delayed drug release and longer drug retention in the pancreas relative to SE-PLGA-427. Diffusion-weighted MRI indicated a consistent decrease in cellularity during drug treatment with both types of drug-loaded nanoparticles. Both SE- and DE-PLGA-427 showed a 6-fold and 4-fold reduction in tumor volume relative to untreated tumors and an elimination of primary pancreatic tumor in 68% of the mice. CONCLUSIONS These results indicated that the PLGA nanoparticles improved drug delivery of PHT-427 to pancreatic tumors, which improved the treatment of MIA PaCa-2 pancreatic cancer.
Collapse
|
5
|
Borisov MA, Petrakova OS, Gvazava IG, Kalistratova EN, Vasiliev AV. Stem Cells in the Treatment of Insulin-Dependent Diabetes Mellitus. Acta Naturae 2016; 8:31-43. [PMID: 27795842 PMCID: PMC5081704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Indexed: 11/02/2022] Open
Abstract
Diabetes affects over 350 million people worldwide, with the figure projected to rise to nearly 500 million over the next 20 years, according to the World Health Organization. Insulin-dependent diabetes mellitus (type 1 diabetes) is an endocrine disorder caused by an autoimmune reaction that destroys insulin-producing β-cells in the pancreas, which leads to insulin deficiency. Administration of exogenous insulin remains at the moment the treatment mainstay. This approach helps to regulate blood glucose levels and significantly increases the life expectancy of patients. However, type 1 diabetes is accompanied by long-term complications associated with the systemic nature of the disease and metabolic abnormalities having a profound impact on health. Of greater impact would be a therapeutic approach which would overcome these limitations by better control of blood glucose levels and prevention of acute and chronic complications. The current efforts in the field of regenerative medicine are aimed at finding such an approach. In this review, we discuss the time-honored technique of donor islets of Langerhans transplantation. We also focus on the use of pluripotent stem and committed cells and cellular reprogramming. The molecular mechanisms of pancreatic differentiation are highlighted. Much attention is devoted to the methods of grafts delivery and to the materials used during its creation.
Collapse
Affiliation(s)
- M. A. Borisov
- Pirogov Russian National Research Medical University, Ostrovitianov str. 1, Moscow, 117997, Russia
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Vavilova str. 26, Moscow, 119334, Russia
| | - O. S. Petrakova
- Pirogov Russian National Research Medical University, Ostrovitianov str. 1, Moscow, 117997, Russia
- Lomonosov Moscow State University, Faculty of Biology, Leninskie Gory 1, bld. 12, Moscow, 119991 , Russia
| | - I. G. Gvazava
- Pirogov Russian National Research Medical University, Ostrovitianov str. 1, Moscow, 117997, Russia
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Vavilova str. 26, Moscow, 119334, Russia
| | - E. N. Kalistratova
- Lomonosov Moscow State University, Faculty of Biology, Leninskie Gory 1, bld. 12, Moscow, 119991 , Russia
| | - A. V. Vasiliev
- Lomonosov Moscow State University, Faculty of Biology, Leninskie Gory 1, bld. 12, Moscow, 119991 , Russia
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Vavilova str. 26, Moscow, 119334, Russia
| |
Collapse
|
6
|
Ding L, Gysemans C, Mathieu C. β-Cell differentiation and regeneration in type 1 diabetes. Diabetes Obes Metab 2013; 15 Suppl 3:98-104. [PMID: 24003926 DOI: 10.1111/dom.12164] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 04/24/2013] [Indexed: 12/15/2022]
Abstract
Pancreatic insulin-producing β-cells have traditionally been viewed as a quiescent cell population. However, several recent lines of evidence indicated that like most tissues the β-cell mass is dynamically regulated with ongoing β-cell regeneration throughout life to replenish lost or damaged β-cells. In type 1 diabetes (T1D), this fine-tuned balance between β-cell death and β-cell renewal in the endocrine pancreas is lost and the deficit in β-cell mass is largely caused by autoimmune-mediated apoptosis. Currently, the concept that a cure for T1D will require both re-establishment of immunological tolerance along with replacement or regeneration of a functional β-cell mass in T1D patients is generally accepted. In this study our current understanding of the events directing β-cell replication, β-cell reprogramming from different cell types and β-cell regeneration is reviewed, in view of the results of various immunomodulatory strategies aiming at blocking autoimmune responses against pancreatic β-cells and at improving β-cell mass and function in subjects with T1D.
Collapse
Affiliation(s)
- L Ding
- Laboratory of Clinical and Experimental Endocrinology, Campus Gasthuisberg O&N1, Faculty of Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | | | | |
Collapse
|
7
|
Ugleholdt R, Pedersen J, Bassi MR, Füchtbauer EM, Jørgensen SM, Kissow HL, Nytofte N, Poulsen SS, Rosenkilde MM, Seino Y, Thams P, Holst PJ, Holst JJ. Transgenic rescue of adipocyte glucose-dependent insulinotropic polypeptide receptor expression restores high fat diet-induced body weight gain. J Biol Chem 2011; 286:44632-45. [PMID: 22027838 DOI: 10.1074/jbc.m111.311779] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The glucose-dependent insulinotropic polypeptide receptor (GIPr) has been implicated in high fat diet-induced obesity and is proposed as an anti-obesity target despite an uncertainty regarding the mechanism of action. To independently investigate the contribution of the insulinotropic effects and the direct effects on adipose tissue, we generated transgenic mice with targeted expression of the human GIPr to white adipose tissue or beta-cells, respectively. These mice were then cross-bred with the GIPr knock-out strain. The central findings of the study are that mice with GIPr expression targeted to adipose tissue have a similar high fat diet -induced body weight gain as control mice, significantly greater than the weight gain in mice with a general ablation of the receptor. Surprisingly, this difference was due to an increase in total lean body mass rather than a gain in total fat mass that was similar between the groups. In contrast, glucose-dependent insulinotropic polypeptide-mediated insulin secretion does not seem to be important for regulation of body weight after high fat feeding. The study supports a role of the adipocyte GIPr in nutrient-dependent regulation of body weight and lean mass, but it does not support a direct and independent role for the adipocyte or beta-cell GIPr in promoting adipogenesis.
Collapse
Affiliation(s)
- Randi Ugleholdt
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Dissanayake D, Gronski MA, Lin A, Elford AR, Ohashi PS. Immunological perspective of self versus tumor antigens: insights from the RIP-gp model. Immunol Rev 2011; 241:164-79. [PMID: 21488897 DOI: 10.1111/j.1600-065x.2011.01014.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Self-reactive T cells in the body are controlled by mechanisms of peripheral tolerance that limit their activation and induction of immune pathology. Our understanding of these mechanisms has been advanced by the use of tissue-specific promoters to express neo-self-antigens. Here, we present findings using the RIP-gp (rat insulin promoter-glycoprotein) transgenic mouse, which expresses the lymphocytic choriomeningitis virus glycoprotein (LCMV-gp) specifically in the pancreatic β islet cells. T cells responsive to this antigen remain ignorant of the LCMV-gp expressed by the islets, and breaking tolerance is dependent upon the maturation status of antigen-presenting cells, the avidity of the T-cell receptor ligation, and the level of major histocompatibility complex expression in the pancreas. Furthermore, decreased activity of Casitas B-lineage lymphoma b, a negative regulator of T-cell receptor signaling, can allow recognition and destruction of the pancreatic islets. This review discusses the roles of these factors in the context of anti-tissue responses, both in the setting of autoimmunity and in anti-tumor immunity.
Collapse
Affiliation(s)
- Dilan Dissanayake
- Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, Toronto, ON, Canada
| | | | | | | | | |
Collapse
|
9
|
Link A, Hardie DL, Favre S, Britschgi MR, Adams DH, Sixt M, Cyster JG, Buckley CD, Luther SA. Association of T-zone reticular networks and conduits with ectopic lymphoid tissues in mice and humans. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:1662-75. [PMID: 21435450 PMCID: PMC3070229 DOI: 10.1016/j.ajpath.2010.12.039] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 12/06/2010] [Accepted: 12/17/2010] [Indexed: 01/08/2023]
Abstract
Ectopic or tertiary lymphoid tissues (TLTs) are often induced at sites of chronic inflammation. They typically contain various hematopoietic cell types, high endothelial venules, and follicular dendritic cells; and are organized in lymph node-like structures. Although fibroblastic stromal cells may play a role in TLT induction and persistence, they have remained poorly defined. Herein, we report that TLTs arising during inflammation in mice and humans in a variety of tissues (eg, pancreas, kidney, liver, and salivary gland) contain stromal cell networks consisting of podoplanin(+) T-zone fibroblastic reticular cells (TRCs), distinct from follicular dendritic cells. Similar to lymph nodes, TRCs were present throughout T-cell-rich areas and had dendritic cells associated with them. They expressed lymphotoxin (LT) β receptor (LTβR), produced CCL21, and formed a functional conduit system. In rat insulin promoter-CXCL13-transgenic pancreas, the maintenance of TRC networks and conduits was partially dependent on LTβR and on lymphoid tissue inducer cells expressing LTβR ligands. In conclusion, TRCs and conduits are hallmarks of secondary lymphoid organs and of well-developed TLTs, in both mice and humans, and are likely to act as important scaffold and organizer cells of the T-cell-rich zone.
Collapse
Affiliation(s)
- Alexander Link
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Chee J, Angstetra E, Mariana L, Graham KL, Carrington EM, Bluethmann H, Santamaria P, Allison J, Kay TWH, Krishnamurthy B, Thomas HE. TNF receptor 1 deficiency increases regulatory T cell function in nonobese diabetic mice. THE JOURNAL OF IMMUNOLOGY 2011; 187:1702-12. [PMID: 21734073 DOI: 10.4049/jimmunol.1100511] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
TNF has been implicated in the pathogenesis of type 1 diabetes. When administered early in life, TNF accelerates and increases diabetes in NOD mice. However, when administered late, TNF decreases diabetes incidence and delays onset. TNFR1-deficient NOD mice were fully protected from diabetes and only showed mild peri-insulitis. To further dissect how TNFR1 deficiency affects type 1 diabetes, these mice were crossed to β cell-specific, highly diabetogenic TCR transgenic I-A(g7)-restricted NOD4.1 mice and Kd-restricted NOD8.3 mice. TNFR1-deficient NOD4.1 and NOD8.3 mice were protected from diabetes and had significantly less insulitis compared with wild type NOD4.1 and NOD8.3 controls. Diabetic NOD4.1 mice rejected TNFR1-deficient islet grafts as efficiently as control islets, confirming that TNFR1 signaling is not directly required for β cell destruction. Flow cytometric analysis showed a significant increase in the number of CD4(+)CD25(+)Foxp3(+) T regulatory cells in TNFR1-deficient mice. TNFR1-deficient T regulatory cells were functionally better at suppressing effector cells than were wild type T regulatory cells both in vitro and in vivo. This study suggests that blocking TNF signaling may be beneficial in increasing the function of T regulatory cells and suppression of type 1 diabetes.
Collapse
Affiliation(s)
- Jonathan Chee
- St. Vincent's Institute, Fitzroy, Victoria 3065, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Desgraz R, Bonal C, Herrera PL. β-cell regeneration: the pancreatic intrinsic faculty. Trends Endocrinol Metab 2011; 22:34-43. [PMID: 21067943 DOI: 10.1016/j.tem.2010.09.004] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 09/27/2010] [Accepted: 09/27/2010] [Indexed: 01/08/2023]
Abstract
Type I diabetes (T1D) patients rely on cumbersome chronic injections of insulin, making the development of alternate durable treatments a priority. The ability of the pancreas to generate new β-cells has been described in experimental diabetes models and, importantly, in infants with T1D. Here we discuss recent advances in identifying the origin of new β-cells after pancreatic injury, with and without inflammation, revealing a surprising degree of cell plasticity in the mature pancreas. In particular, the inducible selective near-total destruction of β-cells in healthy adult mice uncovers the intrinsic capacity of differentiated pancreatic cells to spontaneously reprogram to produce insulin. This opens new therapeutic possibilities because it implies that β-cells can differentiate endogenously, in depleted adults, from heterologous origins.
Collapse
Affiliation(s)
- Renaud Desgraz
- Department of Cell Physiology and Metabolism, University of Geneva Faculty of Medicine, 1 rue Michel-Servet, 1211 Geneva 4, Switzerland
| | | | | |
Collapse
|
12
|
Abstract
Apoptosis of beta cells is a feature of both type 1 and type 2 diabetes as well as loss of islets after transplantation. In type 1 diabetes, beta cells are destroyed by immunological mechanisms. In type 2 diabetes abnormal levels of metabolic factors contribute to beta cell failure and subsequent apoptosis. Loss of beta cells after islet transplantation is due to many factors including the stress associated with islet isolation, primary graft non-function and allogeneic graft rejection. Irrespective of the exact mediators, highly conserved intracellular pathways of apoptosis are triggered. This review will outline the molecular mediators of beta cell apoptosis and the intracellular pathways activated.
Collapse
Affiliation(s)
- Helen E Thomas
- St. Vincent's Institute of Medical Research, 41 Victoria Parade, Fitzroy, VIC 3065, Australia.
| | | | | | | | | |
Collapse
|
13
|
Kobayashi T, Tanaka S, Aida K, Takizawa S, Shimura H, Endo T. Diabetes associated with autoimmune pancreatitis: new insights into the mechanism of β-cell dysfunction. Expert Rev Endocrinol Metab 2009; 4:591-602. [PMID: 30780791 DOI: 10.1586/eem.09.42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
A high proportion of patients with autoimmune pancreatitis (AIP) have diabetes. The decreased β-cell function in active AIP, which leads to diabetes, can sometimes be reversed by corticosteroid treatment. However, the immunological mechanisms causing this β-cell dysfunction are largely unclear. Our recent studies on AIP complicated with diabetes, and data from other animal models of AIP, suggest the presence of distinct mechanisms responsible for β-cell damage in AIP. The presence of immunological cross-reactivity against antigens that are localized both in exocrine pancreatic tissue and β-cells may explain the concomitant occurrence of pancreatitis and β-cell damage in AIP.
Collapse
Affiliation(s)
- Tetsuro Kobayashi
- a Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan.
| | - Shoichiro Tanaka
- b Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan.
| | - Kaoru Aida
- c Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan.
| | - Soichi Takizawa
- d Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan.
| | - Hiroki Shimura
- e Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan.
| | - Toyoshi Endo
- f Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan.
| |
Collapse
|
14
|
Abstract
Prospects for inducing endogenous beta-cell regeneration in the pancreas, one of the most attractive approaches to reverse type 1 and type 2 diabetes, have gained substantially from recent evidence that cells in the adult pancreas exhibit more plasticity than previously recognized. There are two major pathways to beta-cell regeneration, beta-cell replication and beta-cell neogenesis. Substantial evidence for a role for both processes exists in different models. While beta-cell replication clearly occurs during development and early in life, the potential for replication appears to decline substantially with age. In contrast, we have demonstrated that the exocrine compartment of the adult human pancreas contains a facultative stem cell that can differentiate into beta-cells under specific circumstances. We have favoured the idea that, similar to models described in liver regeneration, beta-cell mass can be increased either by neogenesis or replication, depending on the intensity of different stimuli or stressors. Understanding the nature of endocrine stem/progenitor cells and the mechanism by which external stimuli mobilize them to exhibit endocrine differentiation is central for success in therapeutic approaches to induce meaningful endogenous beta-cell neogenesis.
Collapse
Affiliation(s)
- C Demeterco
- Department of Pediatrics, University of California San Diego, Rady Children's Hospital, La Jolla, USA
| | | | | | | | | |
Collapse
|
15
|
Dirice E, Sanlioglu AD, Kahraman S, Ozturk S, Balci MK, Omer A, Griffith TS, Sanlioglu S. Adenovirus-Mediated TRAIL Gene (Ad5hTRAIL) Delivery into Pancreatic Islets Prolongs Normoglycemia in Streptozotocin-Induced Diabetic Rats. Hum Gene Ther 2009; 20:1177-89. [DOI: 10.1089/hum.2009.039] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Ercument Dirice
- Human Gene Therapy Division, Department of Medical Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
- Department of Medical Biology and Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| | - Ahter Dilsad Sanlioglu
- Human Gene Therapy Division, Department of Medical Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
- Department of Medical Biology and Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| | - Sevim Kahraman
- Human Gene Therapy Division, Department of Medical Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
- Department of Medical Biology and Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| | - Saffet Ozturk
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| | - Mustafa Kemal Balci
- Division of Endocrinology and Metabolic Diseases, Department of Medicine, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| | - Abdulkadir Omer
- Section on Islet Transplantation and Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, U.S.A
| | | | - Salih Sanlioglu
- Human Gene Therapy Division, Department of Medical Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
- Department of Medical Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| |
Collapse
|
16
|
Sanlioglu AD, Griffith TS, Omer A, Dirice E, Sari R, Altunbas HA, Balci MK, Sanlioglu S. Molecular mechanisms of death ligand-mediated immune modulation: a gene therapy model to prolong islet survival in type 1 diabetes. J Cell Biochem 2008; 104:710-20. [PMID: 18247339 DOI: 10.1002/jcb.21677] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Type 1 diabetes results from the T cell-mediated destruction of pancreatic beta cells. Islet transplantation has recently become a potential therapeutic approach for patients with type 1 diabetes. However, islet-graft failure appears to be a challenging issue to overcome. Thus, complementary gene therapy strategies are needed to improve the islet-graft survival following transplantation. Immune modulation through gene therapy represents a novel way of attacking cytotoxic T cells targeting pancreatic islets. Various death ligands of the TNF family such as FasL, TNF, and TNF-Related Apoptosis-Inducing Ligand (TRAIL) have been studied for this purpose. The over-expression of TNF or FasL in pancreatic islets exacerbates the onset of type 1 diabetes generating lymphocyte infiltrates responsible for the inflammation. Conversely, the lack of TRAIL expression results in higher degree of islet inflammation in the pancreas. In addition, blocking of TRAIL function using soluble TRAIL receptors facilitates the onset of diabetes. These results suggested that contrary to what was observed with TNF or FasL, adenovirus mediated TRAIL gene delivery into pancreatic islets is expected to be therapeutically beneficial in the setting of experimental models of type 1 diabetes. In conclusion; this study mainly reveals the fundamental principles of death ligand-mediated immune evasion in diabetes mellitus.
Collapse
Affiliation(s)
- Ahter Dilsad Sanlioglu
- Human Gene Therapy Unit and the Department of Medical Biology and Genetics, Akdeniz University, Faculty of Medicine, 07070 Antalya, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Németh ZH, Bleich D, Csóka B, Pacher P, Mabley JG, Himer L, Vizi ES, Deitch EA, Szabó C, Cronstein BN, Haskó G. Adenosine receptor activation ameliorates type 1 diabetes. FASEB J 2007; 21:2379-88. [PMID: 17405852 PMCID: PMC2225539 DOI: 10.1096/fj.07-8213com] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Growing evidence indicates that adenosine receptors could be promising therapeutic targets in autoimmune diseases. Here we studied the role of adenosine receptors in controlling the course of type 1 diabetes. Diabetes in CD-1 mice was induced by multiple-low-dose-streptozotocin (MLDS) treatment and in nonobese diabetic (NOD) mice by cyclophosphamide injection. The nonselective adenosine receptor agonist 5'-N-ethylcarboxamidoadenosine (NECA) prevented diabetes development in both MLDS-challenged mice and in cyclophosphamide-treated NOD mice. The effect of NECA was reversed by the selective A2B receptor antagonist N-(4-cyanophenyl)-2-[4-(2,3,6,7-tetrahydro-2,6-dioxo-1,3-dipropyl-1H-purin-8-yl)phenoxy]acetamide (MRS 1754). The selective A1 receptor agonist 2-chloro-N6-cyclopentyladenosine (CCPA) and A3 receptor agonist N6-(3-iodobenzyl)-adenosine-5'-N-methyluronamide (IB-MECA) were less efficacious in ameliorating the course of diabetes. NECA inhibited diabetes in A2A receptor KO mice and the selective A2A receptor agonist 2-p-(2-carboxyethyl)phenethyl-amino-5'-N-ethyl-carboxamidoadenosine (CGS21680) had no effect in normal mice, indicating a lack of role of A2A receptors. NECA failed to prevent cytokine-induced beta-cell death in vitro, but NECA strongly suppressed expression of the proinflammatory cytokines TNF-alpha, MIP-1alpha, IL-12, and IFN-gamma in pancreata, endotoxin, or anti-CD3-stimulated splenic cells, and T helper 1 lymphocytes, indicating that the beneficial effect of NECA was due to immunomodulation. These results demonstrate that adenosine receptor ligands are potential candidates for the treatment of type 1 diabetes.
Collapse
MESH Headings
- Adenosine-5'-(N-ethylcarboxamide)/pharmacology
- Adenosine-5'-(N-ethylcarboxamide)/therapeutic use
- Animals
- Cells, Cultured/drug effects
- Cells, Cultured/physiology
- Cytokines/metabolism
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Experimental/prevention & control
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/physiopathology
- Diabetes Mellitus, Type 1/prevention & control
- Gene Amplification
- Insulin/metabolism
- Male
- Mice
- Mice, Inbred NOD
- Pancreas/drug effects
- Pancreas/metabolism
- Pancreas/pathology
- Purinergic P1 Receptor Agonists
- Receptors, Purinergic P1/genetics
- Receptors, Purinergic P1/physiology
- Th1 Cells/immunology
- Th2 Cells/immunology
Collapse
Affiliation(s)
- Zoltán H. Németh
- Department of Surgery, University of Medicine and Dentistry of New Jersey, New Jersey Medical School, Newark, New Jersey, USA
| | - David Bleich
- Department of Medicine, University of Medicine and Dentistry of New Jersey, New Jersey Medical School, Newark, New Jersey, USA
| | - Balázs Csóka
- Department of Surgery, University of Medicine and Dentistry of New Jersey, New Jersey Medical School, Newark, New Jersey, USA
| | - Pál Pacher
- Section on Oxidative Stress and Tissue Injury, Laboratory of Physiological Studies, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Jon G. Mabley
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Cockcroft Building, Brighton, UK
| | - Leonóra Himer
- Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Hungary
| | - E. Sylvester Vizi
- Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Hungary
| | - Edwin A. Deitch
- Department of Surgery, University of Medicine and Dentistry of New Jersey, New Jersey Medical School, Newark, New Jersey, USA
| | - Csaba Szabó
- Department of Surgery, University of Medicine and Dentistry of New Jersey, New Jersey Medical School, Newark, New Jersey, USA
| | - Bruce N. Cronstein
- Division of Clinical Pharmacology, Department of Medicine, New York University School of Medicine, New York, New York, USA
| | - György Haskó
- Department of Surgery, University of Medicine and Dentistry of New Jersey, New Jersey Medical School, Newark, New Jersey, USA
- Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Hungary
- Correspondence: Department of Surgery, UMDNJ-New Jersey Medical School, 185 South Orange Ave., University Heights, Newark, NJ 07103, USA. E-mail:
| |
Collapse
|
18
|
TANAKA SHOICHIRO, KOBAYASHI TETSURO, NAKANISHI KOJI, OKUBO MINORU, ODAWARA MASATO, MURASE TOSHIO, HASHIMOTO MASAJI, WATANABE GORO, MATSUSHITA HIROSHI, INOKO HIDETOSHI, TAKEUCHI KAZUO. Corticosteroid-Responsive Diabetes Mellitus Associated with Autoimmune Pancreatitis. Ann N Y Acad Sci 2006. [DOI: 10.1111/j.1749-6632.2002.tb02959.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
19
|
Haase C, Skak K, Michelsen BK, Markholst H. Local activation of dendritic cells leads to insulitis and development of insulin-dependent diabetes in transgenic mice expressing CD154 on the pancreatic beta-cells. Diabetes 2004; 53:2588-95. [PMID: 15448088 DOI: 10.2337/diabetes.53.10.2588] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The initial events leading to activation of the immune system in type 1 diabetes are still largely unknown. In vivo, dendritic cells (DCs) are thought to be the only antigen-presenting cells (APCs) capable of activating naïve T-cells and are therefore important for the initiation of the autoimmune response. To test the effect of activating islet-associated APCs in situ, we generated transgenic mice expressing CD154 (CD40 ligand) under control of the rat insulin promoter (RIP). RIP-CD154 mice developed both insulitis and diabetes, although with different incidence in independent lines. We show that activated DCs could be detected both in the pancreas and in the draining pancreatic lymph nodes. Furthermore, diabetes development was dependent on the presence of T- and B-cells since recombination-activating gene (RAG)-deficient RIP-CD154 mice did not develop diabetes. Finally, we show that the activation of immune cells was confined to the pancreas because transplantation of nontransgenic islets to diabetic recipients restored normoglycemia. Together, these data suggest that expression of CD154 on the beta-cells can lead to activation of islet-associated APCs that will travel to the lymph nodes and activate the immune system, leading to insulitis and diabetes.
Collapse
Affiliation(s)
- Claus Haase
- Hagedorn Research Institute, Niels Steensens Vej 6, DK-2820 Gentofte, Denmark
| | | | | | | |
Collapse
|
20
|
Chen Y, Chong MMW, Darwiche R, Thomas HE, Kay TWH. Severe pancreatitis with exocrine destruction and increased islet neogenesis in mice with suppressor of cytokine signaling-1 deficiency. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 165:913-21. [PMID: 15331415 PMCID: PMC1618606 DOI: 10.1016/s0002-9440(10)63353-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Mice with suppressor of cytokine signaling-1 (SOCS-1) deficiency die within 3 weeks of birth from a multiorgan inflammatory disease. Increased systemic levels and sensitivity of cells to the inflammatory cytokines interferon-gamma and tumor necrosis factor may contribute to the disease. Hepatitis and liver failure are thought to be the cause of the neonatal lethality in these mice. Here, we show that the pancreata of SOCS-1(-/-) mice are also severely affected by inflammation, displaying extensive edema and infiltration by T cells and macrophages. Acinar cells in particular were atrophied and reduced in their zymogen content. The expression of inflammatory markers, including class I major histocompatibility complex and inducible nitric oxide synthase, were increased in the SOCS-1(-/-) pancreas. Although there was generalized up-regulation of class I major histocompatibility complex, inducible nitric oxide synthase expression was more prominent on exocrine tissues. There appeared to be preferential damage and apoptosis of exocrine over endocrine components. Unexpectedly, increased islet neogenesis, possibly from proliferating ductal cells, was observed in the pancreas of SOCS-1(-/-) mice. This is reminiscent of the pancreatitis and islet neogenesis that occur in mice that transgenically overexpress interferon-gamma and/or tumor necrosis factor. This study suggests that in addition to liver failure, the pancreatitis may also be an important contributor to the neonatal lethality in SOCS-1(-/-) mice.
Collapse
Affiliation(s)
- Ye Chen
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | | | | | | | | |
Collapse
|
21
|
Homo-Delarche F, Drexhage HA. Immune cells, pancreas development, regeneration and type 1 diabetes. Trends Immunol 2004; 25:222-9. [PMID: 15099561 DOI: 10.1016/j.it.2004.02.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Françoise Homo-Delarche
- CNRS UMR 7059, Université Paris 7/Denis Diderot, 2 place Jussieu, 75251 Paris Cedex 05, France.
| | | |
Collapse
|
22
|
Sarween N, Chodos A, Raykundalia C, Khan M, Abbas AK, Walker LSK. CD4+CD25+ Cells Controlling a Pathogenic CD4 Response Inhibit Cytokine Differentiation, CXCR-3 Expression, and Tissue Invasion. THE JOURNAL OF IMMUNOLOGY 2004; 173:2942-51. [PMID: 15322152 DOI: 10.4049/jimmunol.173.5.2942] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
It is well established that CD4(+)CD25(+) regulatory T cells (Tregs) inhibit autoimmune pathology. However, precisely how the behavior of disease-inducing T cells is altered by Tregs remains unclear. In this study we use a TCR transgenic model of diabetes to pinpoint how pathogenic CD4 T cells are modified by Tregs in vivo. We show that although Tregs only modestly inhibit CD4 cell expansion, they potently suppress tissue infiltration. This is associated with a failure of CD4 cells to differentiate into effector cells and to up-regulate the IFN-gamma-dependent chemokine receptor CXCR-3, which confers the ability to respond to pancreatic islet-derived CXCL10. Our data support a model in which Tregs permit T cell activation, yet prohibit T cell differentiation and migration into Ag-bearing tissues.
Collapse
Affiliation(s)
- Nadia Sarween
- Medical Research Council Center for Immune Regulation, University of Birmingham Medical School, Birmingham, United Kingdom
| | | | | | | | | | | |
Collapse
|
23
|
Rabinovitch A. Immunoregulation by cytokines in autoimmune diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2003; 520:159-93. [PMID: 12613578 DOI: 10.1007/978-1-4615-0171-8_10] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
24
|
Meagher C, Sharif S, Hussain S, Cameron MJ, Arreaza GA, Delovitch TL. Cytokines and chemokines in the pathogenesis of murine type 1 diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2003; 520:133-58. [PMID: 12613577 DOI: 10.1007/978-1-4615-0171-8_9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Affiliation(s)
- C Meagher
- The Robarts Research Institute and University of Western Ontario, Department of Microbiology and Immunology, and Medicine, London, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
25
|
Kay TWH, Darwiche R, Irawaty W, Chong MMW, Pennington HL, Thomas HE. The role of cytokines as effectors of tissue destruction in autoimmunity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2003; 520:73-86. [PMID: 12613573 DOI: 10.1007/978-1-4615-0171-8_5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Thomas W H Kay
- The Walter and Eliza Hall Institute, Burnet Clinical Research Unit, Parkville, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
26
|
Rajagopalan G, Kudva YC, Flavell RA, David CS. Accelerated diabetes in rat insulin promoter-tumor necrosis factor-alpha transgenic nonobese diabetic mice lacking major histocompatibility class II molecules. Diabetes 2003; 52:342-7. [PMID: 12540606 DOI: 10.2337/diabetes.52.2.342] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The major predisposing genetic component in type 1 diabetes maps to the major histocompatibility complex locus in both mice and humans. To verify the HLA class II association with disease pathogenesis, we adopted the transgenic approach. Expression of HLA-DQ8, the molecule showing the strongest association with human type 1 diabetes, in the diabetes-predisposing milieu of NOD mice in the absence of the endogenous class II molecule I-A(g7) did not render susceptibility to type 1 diabetes. To study if providing a local proinflammatory environment would lead to diabetes in these mice, Abeta(o).NOD.DQ8 were bred with C57BL/6 mice expressing tumor necrosis factor (TNF)-alpha in the beta-cells of the islets of Langerhans. Surprisingly, although diabetes was evident in the F1 intercross expressing rat insulin promoter (RIP)-TNF, offspring lacking either endogenous or transgenic class II molecules developed accelerated diabetes with high frequency in both sexes. Moreover, expression of any functional class II molecule seemed to confer significant protection from diabetes in this model. Thus, neonatal expression of TNF-alpha in an islet-specific manner bypassed the requirement of CD4(+) T-cells and resulted in diabetes that could be mediated by CD8(+) T-cells. We also show for the first time that diabetes in NOD.RIP-TNF mice can occur independent of inheritance of NOD-derived idd1.
Collapse
|
27
|
Ventura-Oliveira D, Vilella CA, Zanin ME, Castro GM, Moreira Filho DC, Zollner RL. Kinetics of TNF-alpha and IFN-gamma mRNA expression in islets and spleen of NOD mice. Braz J Med Biol Res 2002; 35:1347-55. [PMID: 12426635 DOI: 10.1590/s0100-879x2002001100013] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Insulin-dependent diabetes mellitus is caused by autoimmune destruction of pancreatic beta cells. Non-obese diabetic (NOD) mice spontaneously develop diabetes similar to the human disease. Cytokines produced by islet-infiltrating mononuclear cells may be directly cytotoxic and can be involved in islet destruction coordinated by CD4+ and CD8+ cells. We utilized a semiquantitative RT-PCR assay to analyze in vitro the mRNA expression of TNF-alpha and IFN-gamma cytokine genes in isolated islets (N = 100) and spleen cells (5 x 10(5) cells) from female NOD mice during the development of diabetes and from female CBA-j mice as a related control strain that does not develop diabetes. Cytokine mRNAs were measured at 2, 4, 8, 14 and 28 weeks of age from the onset of insulitis to the development of overt diabetes. An increase in IFN-gamma expression in islets was observed for females aged 28 weeks (149 +/- 29 arbitrary units (AU), P<0.05, Student t-test) with advanced destructive insulitis when compared with CBA-j mice, while TNF-alpha was expressed in both NOD and CBA-j female islets at the same level at all ages studied. In contrast, TNF-alpha in spleen was expressed at higher levels in NOD females at 14 weeks (99 +/- 8 AU, P<0.05) and 28 weeks (144 +/- 17 AU, P<0.05) of age when compared to CBA-j mice. The data suggest that IFN-gamma and TNF-alpha expression in pancreatic islets of female NOD mice is associated with beta cell destruction and overt diabetes.
Collapse
Affiliation(s)
- D Ventura-Oliveira
- Disciplina e Laboratório de Imunologia Clínica, Departamento de Clínica Médica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, SP, Brasil
| | | | | | | | | | | |
Collapse
|
28
|
Kollias G, Kontoyiannis D. Role of TNF/TNFR in autoimmunity: specific TNF receptor blockade may be advantageous to anti-TNF treatments. Cytokine Growth Factor Rev 2002; 13:315-21. [PMID: 12220546 DOI: 10.1016/s1359-6101(02)00019-9] [Citation(s) in RCA: 146] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Deregulated TNF production, be it low or high, characterizes many autoimmune diseases. Recent evidence supports a dualistic, pro-inflammatory and immune- or disease-suppressive role for TNF in these conditions. Blocking TNF in autoimmune-prone chronic inflammatory diseases may, therefore, lead to unpredictable outcomes, depending on timing and duration of treatment. Indeed, blockade of TNF in human rheumatoid arthritis or inflammatory bowel disease patients, although so far impressively beneficial for the majority of patients, it has also led to a significant incidence of drug induced anti-dsDNA production or even in manifestations of lupus and neuro-inflammatory disease. Notably, anti-TNF treatment of multiple sclerosis patients has led almost exclusively to immune activation and disease exacerbation. We discuss here recent evidence in murine disease models, indicating an heterogeneity of TNF receptor usage in autoimmune suppression versus inflammatory tissue damage, and put forward a rationale for a predictably beneficial effect of 'anti-TNFR' instead of 'anti-TNF' treatment in human chronic inflammatory and autoimmune conditions.
Collapse
MESH Headings
- Animals
- Antigens, CD/physiology
- Apoptosis/drug effects
- Apoptosis/physiology
- Autoimmune Diseases/drug therapy
- Autoimmune Diseases/immunology
- Autoimmune Diseases/physiopathology
- Autoimmunity/physiology
- Cell Differentiation/drug effects
- Cell Differentiation/physiology
- Crosses, Genetic
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Humans
- Immune Tolerance/physiology
- Inflammation/physiopathology
- Lupus Erythematosus, Systemic/drug therapy
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/physiopathology
- Lymphocyte Subsets/immunology
- Mice
- Mice, Inbred MRL lpr
- Mice, Inbred NOD
- Mice, Inbred NZB
- Mice, Transgenic
- Myelin Sheath/immunology
- Receptors, Tumor Necrosis Factor/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor/physiology
- Receptors, Tumor Necrosis Factor, Type I
- Receptors, Tumor Necrosis Factor, Type II
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
- Tumor Necrosis Factor-alpha/deficiency
- Tumor Necrosis Factor-alpha/physiology
Collapse
Affiliation(s)
- George Kollias
- Biomedical Sciences Research Centre, Institute for Immunology, Alexander Fleming, 14-16 Alexander Fleming Street, 166-72 Vari, Athens, Greece.
| | | |
Collapse
|
29
|
Mori R, Kondo T, Ohshima T, Ishida Y, Mukaida N. Accelerated wound healing in tumor necrosis factor receptor p55-deficient mice with reduced leukocyte infiltration. FASEB J 2002; 16:963-74. [PMID: 12087057 DOI: 10.1096/fj.01-0776com] [Citation(s) in RCA: 211] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
To clarify biological roles of tumor necrosis factor receptor p55 (TNF-Rp55) -mediated signals in wound healing, skin excisions were prepared in BALB/c (WT) and TNF-Rp55-deficient (KO) mice. In WT mice, the wound area was reduced to 50% of the original area 6 days after injury, with angiogenesis and collagen accumulation. Histopathologically, reepithelialization rate was approximately 80% 6 days. Myeloperoxidase activity and macrophage recruitment were the most evident 1 and 6 days after injury, respectively. Gene expression of adhesion molecules, interleukin 1alpha (IL-1alpha), IL-1beta, monocyte chemoattractant protein 1, macrophage inflammatory protein 1alpha (MIP-1alpha), MIP-2, transforming growth factor beta1 (TGF-beta1) connective tissue growth factor (CTGF), vascular endothelial growth factor (VEGF), Flt-1, and Flk-1 was enhanced at the wound site. In KO mice, an enhancement in angiogenesis, collagen content, and reepithelialization was accelerated with the increased gene expression of TGF-beta1, CTGF, VEGF, Flt-1, and Flk-1 at the wound sites, resulting in accelerated wound healing compared with WT mice. In contrast, leukocyte infiltration, mRNA expression of adhesion molecules, and cytokines were significantly reduced in KO mice. These observations suggest that TNF-Rp55-mediated signals have some role in promoting leukocyte infiltration at the wound site and negatively affect wound healing, probably by reducing angiogenesis and collagen accumulation.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, CD/physiology
- Cell Adhesion Molecules/biosynthesis
- Cell Adhesion Molecules/genetics
- Cell Movement
- Cytokines/biosynthesis
- Cytokines/genetics
- Epidermis/anatomy & histology
- Epidermis/physiology
- Growth Substances/biosynthesis
- Growth Substances/genetics
- Hydroxyproline/analysis
- Kinetics
- Leukocytes/immunology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Neovascularization, Physiologic
- Peroxidase/metabolism
- RNA, Messenger/biosynthesis
- Receptor Protein-Tyrosine Kinases/biosynthesis
- Receptor Protein-Tyrosine Kinases/genetics
- Receptors, Growth Factor/biosynthesis
- Receptors, Growth Factor/genetics
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor/physiology
- Receptors, Tumor Necrosis Factor, Type I
- Receptors, Tumor Necrosis Factor, Type II
- Receptors, Vascular Endothelial Growth Factor
- Skin/anatomy & histology
- Skin/blood supply
- Skin/immunology
- Skin Physiological Phenomena/immunology
- Tumor Necrosis Factor-alpha/biosynthesis
- Tumor Necrosis Factor-alpha/genetics
- Wound Healing/immunology
Collapse
Affiliation(s)
- Ryoichi Mori
- Division of Environmental Science, Forensic and Social Environmental Medicine, Graduate School of Medical Science, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | | | | | | | | |
Collapse
|
30
|
Kim JY, Kim YH, Chang I, Kim S, Pak YK, Oh BH, Yagita H, Jung YK, Oh YJ, Lee MS. Resistance of mitochondrial DNA-deficient cells to TRAIL: role of Bax in TRAIL-induced apoptosis. Oncogene 2002; 21:3139-48. [PMID: 12082629 DOI: 10.1038/sj.onc.1205406] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2002] [Revised: 02/06/2002] [Accepted: 02/19/2002] [Indexed: 01/09/2023]
Abstract
Mitochondrion is one of the master players in both apoptosis and necrosis. We studied the role of mitochondrial function in TRAIL-induced apoptosis. TRAIL killed SK-Hep1 cells with characteristic features of apoptosis such as DNA fragmentation, sub-G1 ploidy peak and cytochrome c translocation. In contrast, mitochondrial DNA-deficient SK-Hep1 rho(0) cells were resistant to TRAIL. Dissipation of mitochondrial potential or cytochrome c translocation did not occur in rho(0) cells after TRAIL treatment. TRAIL induced translocation of Bax subsequent to the cleavage of Bid in parental cells. However, Bax translocation was absent in rho(0) cells, accounting for the failure of cytochrome c release in rho(0) cells. Forced expression of Bax induced caspase-3 activity in rho(0) cells. Incubation of rho(0) cells with ADP+Pi to increase intracellular ATP restored sensitivity to TRAIL. Despite different sensitivity to TRAIL, parental cells and rho(0) cells did not show significant difference in susceptibility to agonistic anti-Fas antibody, TNF-alpha or staurosporine. Our results indicate that TRAIL-induced apoptosis is dependent on intact mitochondrial function and susceptibility of mitochondrial DNA-deficient cells to apoptosis depends on the type of apoptotic stimuli. Tumor cells with mitochondrial mutations or dysfunction might have the ability to evade tumor surveillance imposed by TRAIL in vivo.
Collapse
Affiliation(s)
- Ja-Young Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-dong Kangnam-ku, Seoul 135-710, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kitamura K, Nakamoto Y, Akiyama M, Fujii C, Kondo T, Kobayashi K, Kaneko S, Mukaida N. Pathogenic roles of tumor necrosis factor receptor p55-mediated signals in dimethylnitrosamine-induced murine liver fibrosis. J Transl Med 2002; 82:571-83. [PMID: 12003998 DOI: 10.1038/labinvest.3780452] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
TNF-alpha has pleiotropic functions, but its role in liver fibrosis has not yet been clarified. To understand the pathophysiologic role of the TNF-alpha/TNF receptor (TNFR) p55 signals in liver fibrosis, 10 mg/kg of dimethylnitrosamine, a specific hepatotoxicant, was administered twice a week into the peritoneal cavity of both TNFRp55 knock-out (KO) and wild-type mice, and the severity of fibrosis was monitored histologically and biochemically. In wild-type mice, histologic analysis demonstrated evident fibrotic changes 1 week after the initiation of dimethylnitrosamine administration, consistent with increased liver collagen contents. Concomitantly, the numbers of Kupffer cells and activated hepatic stellate cells (HSCs) were increased in liver tissue. On the contrary, fibrotic changes were attenuated and the numbers of Kupffer cells and HSCs were decreased in TNFRp55-KO mice. Moreover, gene expression of TNF-alpha and monocyte chemoattractant protein-1, which are involved in Kupffer cell activation or migration, was decreased in the liver of TNFRp55-KO mice. Collectively, TNFRp55-mediated signals may regulate activation of Kupffer cells and HSCs and eventually enhance fibrotic process.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Collagen/drug effects
- Collagen/metabolism
- DNA Primers/chemistry
- Dimethylnitrosamine/toxicity
- Female
- Gene Expression/drug effects
- Kupffer Cells/metabolism
- Kupffer Cells/pathology
- Liver/drug effects
- Liver/metabolism
- Liver/pathology
- Liver Cirrhosis, Experimental/genetics
- Liver Cirrhosis, Experimental/metabolism
- Liver Cirrhosis, Experimental/pathology
- Matrix Metalloproteinases/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- RNA, Messenger/analysis
- Receptors, Tumor Necrosis Factor/deficiency
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor, Type I
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Specific Pathogen-Free Organisms
- Tissue Inhibitor of Metalloproteinases/metabolism
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Kazuya Kitamura
- Department of Gastroenterology, Graduate School of Medicinal Sciences, Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Tanaka S, Kobayashi T, Nakanishi K, Okubo M, Murase T, Hashimoto M, Watanabe G, Matsushita H, Endo Y, Yoshizaki H, Kosuge T, Sakamoto M, Takeuchi K. Evidence of primary beta-cell destruction by T-cells and beta-cell differentiation from pancreatic ductal cells in diabetes associated with active autoimmune chronic pancreatitis. Diabetes Care 2001; 24:1661-7. [PMID: 11522716 DOI: 10.2337/diacare.24.9.1661] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Diabetes associated with autoimmune chronic pancreatitis (ACP) is a subtype of diabetes that is responsive to corticosteroid treatment of progressive endocrine and exocrine dysfunction. However, little is known about pathological changes of islet and exocrine pancreas in ACP. RESEARCH DESIGN AND METHODS We examined pancreatic specimens obtained on biopsy from four diabetic men with ACP (mean [range]: age 62 years [48-78], duration of ACP 3 months [1-5], duration of diabetes 1 month [0-3]) morphologically, immunohistochemically, and morphometrically. RESULTS The pancreatic specimens in all cases exhibited inflammatory cell infiltration surrounding ductal cells and extensive fibrosis. Some islets were infiltrated with mononuclear cells with disrupted beta-cells. The subsets of T-cells infiltrated to the islets were mainly CD8(+). Islet beta-cell volume was decreased; the mean percentage area of beta-cells in the islets in four cases with ACP were 16% (range 13-20) (P = 0.0015 vs. type 2 diabetic patients, 48% [27-73], n = 8; P = 0.0002 vs. nondiabetic control subjects, 58% [39-77], n = 7). Preserved ductal cells were surrounded predominantly by CD8(+) or CD4(+) T-cells. Some cytokeratin 19-positive ductal cells contained insulin and glucagon, representing upregulated differentiation of islet cells from ductal cells. Insulin promoter factor-1 (IPF-1) was hyperexpressed in insulin-containing ductal cells. CONCLUSIONS Diabetes associated with ACP is caused by T-cell-mediated mechanisms primarily involving islet beta-cells as well as pancreatic ductal cells. In ACP, ductal islet precursor cells were associated with IPF-1 hyperexpression, suggesting a critical role of IPF-1 on islet cell differentiation and eventual beta-cell restoration.
Collapse
Affiliation(s)
- S Tanaka
- Department of Endocrinology and Metabolism, Toranomon Hospital, 2-2-2, Toranomon, Minato-ku, Tokyo 105-8470, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Pakala SV, Ilic A, Chen L, Sarvetnick N. TNF-alpha receptor 1 (p55) on islets is necessary for the expression of LIGHT on diabetogenic T cells. Clin Immunol 2001; 100:198-207. [PMID: 11465949 DOI: 10.1006/clim.2001.5059] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Insulin-dependent diabetes mellitus results from T-cell-mediated destruction of pancreatic islet beta cells. Both CD4 and CD8 T cells have been shown to be independently capable of beta cell destruction. However, the mechanism of beta cell destruction has remained elusive. It has previously been shown that the absence of TNF-alpha receptor 1 (p55) on the islets protected islets from CD4 T-cell-mediated destruction as long as the T cells did not have access to wild-type islets in vivo. Wild-type and TNF-alpha receptor 1 (p55) deficient islets induce similar levels of proliferation of BDC2.5 T cells. In this study, we demonstrate that islet TNF-alpha receptor 1 (p55) influences the expression of LIGHT (TNFSF-14), a TNF family member with both cytolytic and costimulatory properties, on BDC2.5 T cells and the expression of its receptor HVEM (TNFRSF-14) by islets, indicating a role for LIGHT-HVEM interactions in autoimmune diabetes.
Collapse
Affiliation(s)
- S V Pakala
- Department of Immunology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | | | | | | |
Collapse
|
34
|
Cain DM, Khasabov SG, Simone DA. Response properties of mechanoreceptors and nociceptors in mouse glabrous skin: an in vivo study. J Neurophysiol 2001; 85:1561-74. [PMID: 11287480 DOI: 10.1152/jn.2001.85.4.1561] [Citation(s) in RCA: 150] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The increasing use of transgenic mice for the study of pain mechanisms necessitates comprehensive understanding of the murine somatosensory system. Using an in vivo mouse preparation, we studied response properties of tibial nerve afferent fibers innervating glabrous skin. Recordings were obtained from 225 fibers identified by mechanical stimulation of the skin. Of these, 106 were classed as A beta mechanoreceptors, 51 as A delta fibers, and 68 as C fibers. A beta mechanoreceptors had a mean conduction velocity of 22.2 +/- 0.7 (SE) m/s (13.8--40.0 m/s) and a median mechanical threshold of 2.1 mN (0.4--56.6 mN) and were subclassed as rapidly adapting (RA, n = 75) or slowly adapting (SA, n = 31) based on responses to constant force mechanical stimuli. Conduction velocities ranged from 1.4 to 13.6 m/s (mean 7.1 +/- 0.6 m/s) for A delta fibers and 0.21 to 1.3 m/s (0.7 +/- 0.1 m/s) for C fibers. Median mechanical thresholds were 10.4 and 24.4 mN for A delta and C fibers, respectively. Responses of A delta and C fibers evoked by heat (35--51 degrees C) and by cold (28 to -12 degrees C) stimuli were determined. Mean response thresholds of A delta fibers were 42.0 +/- 3.1 degrees C for heat and 7.6 +/- 3.8 degrees C for cold, whereas mean response thresholds of C fibers were 40.3 +/- 0.4 degrees C for heat and 10.1 +/- 1.9 degrees C for cold. Responses evoked by heat and cold stimuli increased monotonically with stimulus intensity. Although only 12% of tested A delta fibers were heat sensitive, 50% responded to cold. Only one A delta nociceptor responded to both heat and cold stimuli. In addition, 40% of A delta fibers were only mechanosensitive since they responded neither to heat nor to cold stimuli. Thermal stimuli evoked responses from the majority of C fibers: 82% were heat sensitive, while 77% of C fibers were excited by cold, and 68% were excited by both heat and cold stimuli. Only 11% of C fibers were insensitive to heat and/or cold. This in vivo study provides an analysis of mouse primary afferent fibers innervating glabrous skin including new information on encoding of noxious thermal stimuli within the peripheral somatosensory system of the mouse. These results will be useful for future comparative studies with transgenic mice.
Collapse
Affiliation(s)
- D M Cain
- Department of Preventive Sciences, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | |
Collapse
|
35
|
Pechhold K, Patterson NB, Blum C, Fleischacker CL, Boehm BO, Harlan DM. Low dose streptozotocin-induced diabetes in rat insulin promoter-mCD80-transgenic mice is T cell autoantigen-specific and CD28 dependent. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:2531-9. [PMID: 11160314 DOI: 10.4049/jimmunol.166.4.2531] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although transgenic mice expressing murine B7-1 (mCD80) on their pancreatic beta cells under the rat insulin-1 promoter (RIP-mCD80(+) mice) rarely develop spontaneous beta cell destruction and diabetes, we have previously reported the transgene-dependent induction of profound insulitis and lethal diabetes following multiple low dose injections of the beta cell toxin streptozotocin (MLDS) in RIP-mCD80(+) mice. Here, we have further characterized this MLDS-induced diabetes model using the RIP-mCD80(+) mice and now demonstrate that disease is critically dependent on T cell signaling via CD28. Thus, although naive RIP-mCD80(+) and nontransgenic littermates have comparable gross beta cell mass, and immediately following MLDS induction the mice display similar degrees of insulitis and decrements in the beta cell mass, only transgenic mice continued to destroy their beta cells and develop insulin-dependent diabetes mellitus. Strikingly, MLDS-induced diabetes was completely prevented in CD28-deficient mice (RIP-mCD80(+)CD28(-/-)) due to abrogation of leukocytes infiltrating their pancreatic islets. We further characterized MLDS-induced diabetes in the RIP-mCD80(+) mice by demonstrating that the MLDS-induced lymphocytic islet infiltrate contained a substantial frequency of autoantigen-specific, IFN-gamma-secreting, CD8(+) T cells. We conclude that MLDS-induced beta cell destruction and subsequent insulin-dependent diabetes mellitus in RIP-mCD80(+) mice is T cell-mediated as it involves both Ag-specific recognition of self-target molecules in the inflamed pancreatic islet (signal 1) and is CD28 costimulation dependent (signal 2).
Collapse
Affiliation(s)
- K Pechhold
- National Institute of Diabetes and Digestive and Kidney Diseases-Navy Transplantation and Autoimmunity Branch, Bethesda, MD 20889, USA
| | | | | | | | | | | |
Collapse
|
36
|
Abraham RS, Wen L, Marietta EV, David CS. Type 1 diabetes-predisposing MHC alleles influence the selection of glutamic acid decarboxylase (GAD) 65-specific T cells in a transgenic model. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:1370-9. [PMID: 11145722 DOI: 10.4049/jimmunol.166.2.1370] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The genetic factors that contribute to the etiology of type 1 diabetes are still largely uncharacterized. However, the genes of the MHC (HLA in humans) have been consistently associated with susceptibility to disease. We have used several transgenic mice generated in our laboratory, bearing susceptible or resistant HLA alleles, in the absence of endogenous MHC class II (Abetao), to study immune responses to the autoantigen glutamic acid decarboxylase (GAD) 65 and its relevance in determining the association between autoreactivity and disease pathogenesis. Mice bearing diabetes-susceptible haplotypes, HLA DR3 (DRB1*0301) or DQ8 (DQB1*0302), singly or in combination showed spontaneous T cell reactivity to rat GAD 65, which is highly homologous to the self Ag, mouse GAD 65. The presence of diabetes-resistant or neutral alleles, such as HLA DQ6 (DQB1*0602) and DR2 (DRB1*1502) prevented the generation of any self-reactive responses to rat GAD. In addition, unmanipulated Abetao/DR3, Abetao/DQ8, and Abetao/DR3/DQ8 mice recognized specific peptides, mainly from the N-terminal region of the GAD 65 molecule. Most of these regions are conserved between human, mouse, and rat GAD 65. Further analysis revealed that the reactivity was mediated primarily by CD4(+) T cells. Stimulation of these T cells by rat GAD 65 resulted in the generation of a mixed Th1/Th2 cytokine profile in the Abetao/DR3/DQ8, Abetao/DR3, and Abetao/DQ8 mice. Thus, the presence of diabetes-associated genes determines whether immune tolerance is maintained to islet autoantigens, but autoreactivity in itself is not sufficient to induce diabetes.
Collapse
Affiliation(s)
- R S Abraham
- Department of Immunology, Mayo Clinic, Rochester, MN 55905. Department of Endocrinology, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | |
Collapse
|
37
|
Toivonen A, Kulmala P, Savola K, Akerblom HK, Knip M. Soluble adhesion molecules in preclinical type 1 diabetes. The Childhood Diabetes in Finland Study Group. Pediatr Res 2001; 49:24-9. [PMID: 11134487 DOI: 10.1203/00006450-200101000-00009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
We measured the concentrations of the soluble forms of the intercellular adhesion molecule-1 (sICAM-1) and L-selectin in 95 autoantibody-positive siblings of children with type 1 diabetes and 95 sex- and age-matched siblings testing negative for diabetes-associated autoantibodies to assess the possible role of soluble adhesion molecules as markers of progressive ss-cell destruction in preclinical diabetes and their ability to discriminate between those siblings who progress to clinical disease and those who remain nondiabetic. We observed an inverse correlation between age and the levels of both sICAM-1 (r = -0.31, p < 0.001) and sL-selectin (r = -0.27, p < 0.001) in the control siblings but no association with HLA-DR phenotypes. There was no difference in the circulating levels of soluble adhesion molecules between the antibody-positive and negative siblings. Among the antibody-positive siblings, those with at least three autoantibodies had higher sICAM-1 levels (p < 0.01) than those testing positive for only one, and siblings with three autoantibodies or more had higher concentrations of sL-selectin (p < 0.01) than those with two autoantibodies. Siblings with an islet cell antibody level of 20 Juvenile Diabetes Foundation units or more had higher sICAM-1 concentrations than those with a level below 20 (p < 0.001), and those testing positive for antibodies to the protein tyrosine phosphatase-related IA-2 antigen had increased levels of both sICAM-1 (p = 0.03) and sL-selectin (p = 0.02) compared with siblings who tested negative. The antibody-positive siblings who progressed to clinical type 1 diabetes were significantly younger than the nonprogressors (p < 0.001) and had higher levels of sICAM-1 initially (p < 0.001). The difference in sICAM-1 concentrations remained significant (p = 0.03) after age adjustment. Our results indicate that concentrations of soluble adhesion molecules are increased in the autoantibody-positive siblings who have the highest risk of developing clinical diabetes, suggesting that ss-cell destruction is reflected in increased circulating levels of these molecules. This is supported by the observation of elevated sICAM-1 concentrations in the 29 siblings who actually progressed to clinical type 1 diabetes. Peripheral levels of soluble adhesion molecules are not able to discriminate between progressors and nonprogressors, however, due to substantial overlapping between these two groups.
Collapse
Affiliation(s)
- A Toivonen
- Medical School, University of Tampere, and the Department of Pediatrics, Tampere University Hospital, FIN-33014 Tampere, Finland
| | | | | | | | | |
Collapse
|
38
|
Abrams JR, Kelley SL, Hayes E, Kikuchi T, Brown MJ, Kang S, Lebwohl MG, Guzzo CA, Jegasothy BV, Linsley PS, Krueger JG. Blockade of T lymphocyte costimulation with cytotoxic T lymphocyte-associated antigen 4-immunoglobulin (CTLA4Ig) reverses the cellular pathology of psoriatic plaques, including the activation of keratinocytes, dendritic cells, and endothelial cells. J Exp Med 2000; 192:681-94. [PMID: 10974034 PMCID: PMC2193278 DOI: 10.1084/jem.192.5.681] [Citation(s) in RCA: 208] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Efficient T cell activation is dependent on the intimate contact between antigen-presenting cells (APCs) and T cells. The engagement of the B7 family of molecules on APCs with CD28 and CD152 (cytotoxic T lymphocyte-associated antigen 4 [CTLA-4]) receptors on T cells delivers costimulatory signal(s) important in T cell activation. We investigated the dependence of pathologic cellular activation in psoriatic plaques on B7-mediated T cell costimulation. Patients with psoriasis vulgaris received four intravenous infusions of the soluble chimeric protein CTLA4Ig (BMS-188667) in a 26-wk, phase I, open label dose escalation study. Clinical improvement was associated with reduced cellular activation of lesional T cells, keratinocytes, dendritic cells (DCs), and vascular endothelium. Expression of CD40, CD54, and major histocompatibility complex (MHC) class II HLA-DR antigens by lesional keratinocytes was markedly reduced in serial biopsy specimens. Concurrent reductions in B7-1 (CD80), B7-2 (CD86), CD40, MHC class II, CD83, DC-lysosomal-associated membrane glycoprotein (DC-LAMP), and CD11c expression were detected on lesional DCs, which also decreased in number within lesional biopsies. Skin explant experiments suggested that these alterations in activated or mature DCs were not the result of direct toxicity of CTLA4Ig for DCs. Decreased lesional vascular ectasia and tortuosity were also observed and were accompanied by reduced presence of E-selectin, P-selectin, and CD54 on vascular endothelium. This study highlights the critical and proximal role of T cell activation through the B7-CD28/CD152 costimulatory pathway in maintaining the pathology of psoriasis, including the newly recognized accumulation of mature DCs in the epidermis.
Collapse
Affiliation(s)
- J R Abrams
- Bristol-Myers Squibb Pharmaceutical Research Institute, Wallingford, Connecticut 06492, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Yamakawa M, Weinstein R, Tsuji T, McBride J, Wong DT, Login GR. Age-related alterations in IL-1beta, TNF-alpha, and IL-6 concentrations in parotid acinar cells from BALB/c and non-obese diabetic mice. J Histochem Cytochem 2000; 48:1033-42. [PMID: 10898798 DOI: 10.1177/002215540004800802] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
IL-1beta, TNF-alpha, and IL-6 have been implicated in the destruction of parotid gland acinar cells (but not duct cells) in autoimmune sialoadenitis. Here we report the temporal alterations of these cytokines in parotid acinar cells that may lead to this specificity in cell death in the non-obese diabetic (NOD) mouse model for Sjögren's syndrome. Immunohistochemistry on paraffin sections of parotid gland from 5- and 10-week-old BALB/c and NOD mice confirmed the presence of many peri-acinar lymphoid nodules but few T-cells and macrophages between acinar cells. RT-PCR on enzymatically dispersed mouse parotid acinar cells (MPACs) showed no bands for CD3varepsilon, CD20, or F4/80 regardless of mouse strain or age. By ELISA, MPACs from 10-week-old NODs showed a small but highly significant (p<0.003) increase in IL-1beta and a large significant decrease (p<0.008) in IL-6 compared to 5-week-old NODs. Norepinephrine-stimulated amylase release from MPACs was not different regardless of mouse strain or age. These data show that alterations in acinar cell production of IL-1beta and IL-6 in aging NODs precede periductal lymphoid aggregates and acinar cell secretory dysfunction. (J Histochem Cytochem 48:1033-1041,2000)
Collapse
Affiliation(s)
- M Yamakawa
- Department of Oral Medicine and Diagnostic Sciences, Harvard School of Dental Medicine, Boston, Massachusetts
| | | | | | | | | | | |
Collapse
|
40
|
Chaturvedi P, Agrawal B, Zechel M, Lee-Chan E, Singh B. A self MHC class II beta-chain peptide prevents diabetes in nonobese diabetic mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:6610-20. [PMID: 10843721 DOI: 10.4049/jimmunol.164.12.6610] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We explored T cell responses to the self class II MHC (I-Ag7) beta-chain-derived peptides in diabetic and prediabetic nonobese diabetic (NOD) mice. We found that one of these immunodominant epitopes of the beta-chain of I-Ag7 molecule, peptide 54-76, could regulate autoimmunity leading to diabetes in NOD mice. T cells from prediabetic young NOD mice do not respond to the peptide 54-76, but T cells from diabetic NOD mice proliferated in response to this peptide. T cells from older nondiabetic mice or mice protected from diabetes do not respond to this peptide, suggesting a role for peptide 54-76-specific T cells in pathogenesis of diabetes. We show that this peptide is naturally processed and presented by the NOD APCs to self T cells. However, the peptide-specific T cells generated after immunization of young mice regulate autoimmunity in NOD mice by blocking the diabetogenic cells in adoptive transfer experiments. The NOD mice immunized with this peptide are protected from both spontaneous and cyclophosphamide-induced insulin-dependent diabetes mellitus. Immunization of young NOD mice with this peptide elicited T cell proliferation and production of Th2-type cytokines. In addition, immunization with this peptide induced peptide-specific Abs of IgG1 isotype that recognized native I-Ag7 molecule on the cell surface and inhibited the T cell proliferative responses. These results suggest that I-Abetag7(54-76) peptide-reactive T cells are involved in the pathogenesis of diabetes. However, immunization with this peptide at young age induces regulatory cells and the peptide-specific Abs that can modulate autoimmunity in NOD mice and prevent spontaneous and induced diabetes.
Collapse
Affiliation(s)
- P Chaturvedi
- Department of Microbiology and Immunology and John P. Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | | | | | | | | |
Collapse
|
41
|
Amrani A, Verdaguer J, Thiessen S, Bou S, Santamaria P. IL-1alpha, IL-1beta, and IFN-gamma mark beta cells for Fas-dependent destruction by diabetogenic CD4(+) T lymphocytes. J Clin Invest 2000; 105:459-68. [PMID: 10683375 PMCID: PMC289158 DOI: 10.1172/jci8185] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Cytokines such as IL-1alpha, IL-1beta, and IFN-gamma have long been implicated in the pathogenesis of autoimmune diabetes, but the mechanisms through which they promote diabetogenesis remain unclear. Here we show that CD4(+) T lymphocytes propagated from transgenic nonobese diabetic (NOD) mice expressing the highly diabetogenic, beta cell-specific 4.1-T-cell receptor (4.1-TCR) can kill IL-1alpha-, IL-1beta-, and IFN-gamma-treated beta cells from NOD mice. Untreated NOD beta cells and cytokine-treated beta cells from Fas-deficient NOD.lpr mice are not targeted by these T cells. Killing of islet cells in vitro was associated with cytokine-induced upregulation of Fas on islet cells and was independent of MHC class II expression. Abrogation of Fas expression in 4.1-TCR-transgenic NOD mice afforded nearly complete protection from diabetes and did not interfere with the development of the transgenic CD4(+) T cells or with their ability to cause insulitis. In contrast, abrogation of perforin expression did not affect beta cell-specific cytotoxicity or the diabetogenic potential of these T cells. These data demonstrate a novel mechanism of action of IL-1alpha, IL-1beta, and IFN-gamma in autoimmune diabetes, whereby these cytokines mark beta cells for Fas-dependent lysis by autoreactive CD4(+) T cells.
Collapse
Affiliation(s)
- A Amrani
- Department of Microbiology, The University of Calgary, Faculty of Medicine, Calgary, Alberta, Canada T2N 4N1
| | | | | | | | | |
Collapse
|
42
|
Herrera PL, Harlan DM, Vassalli P. A mouse CD8 T cell-mediated acute autoimmune diabetes independent of the perforin and Fas cytotoxic pathways: possible role of membrane TNF. Proc Natl Acad Sci U S A 2000; 97:279-84. [PMID: 10618409 PMCID: PMC26654 DOI: 10.1073/pnas.97.1.279] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Double transgenic mice [rat insulin promoter (RIP)-tumor necrosis factor (TNF) and RIP-CD80] whose pancreatic beta cells release TNF and bear CD80 all develop an acute early (6 wk) and lethal diabetes mediated by CD8 T cells. The first ultrastructural changes observed in beta cells, so far unreported, are focal lesions of endoplasmic reticulum swelling at the points of contact with islet-infiltrating lymphoblasts, followed by cytoplasmic, but not nuclear, apoptosis. Such double transgenic mice were made defective in either the perforin, Fas, or TNF pathways. Remarkably, diabetes was found to be totally independent of perforin and Fas. Mice lacking TNF receptor (TNFR) II had no or late diabetes, but only a minority had severe insulitis. Mice lacking the TNF-lymphotoxin (LTalpha) locus (whose sole source of TNF are the beta cells) all had insulitis comparable to that of nondefective mice, but no diabetes or a retarded and milder form, with lesions suggesting different mechanisms of injury. Because both TNFR II and TNF-LTalpha mutations have complex effects on the immune system, these data do not formally incriminate membrane TNF as the major T cell mediator of this acute autoimmune diabetes; nevertheless, in the absence of involvement of the perforin or Fas cytotoxic pathways, membrane TNF appears to be the likeliest candidate.
Collapse
Affiliation(s)
- P L Herrera
- Department of Morphology, University of Geneva Medical School, 1 rue Michel-Servet, 1211 Geneva 4, Switzerland.
| | | | | |
Collapse
|
43
|
Hirai H, Kaino Y, Ito T, Kida K. Analysis of cytokine mRNA expression in pancreatic islets of nonobese diabetic mice. J Pediatr Endocrinol Metab 2000; 13:91-8. [PMID: 10689643 DOI: 10.1515/jpem.2000.13.1.91] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Nonobese diabetic mice develop type 1 diabetes in an age-related and gender-dependent manner. Th1 (IFN-gamma and TNF-beta) and Th2 (IL-4 and IL-10) cytokine mRNA expression was analyzed in pancreatic islets isolated from female NOD mice with a high incidence of diabetes and male NOD mice with a low incidence of diabetes. The levels were measured at 5 time points from the onset of insulitis until the development of overt diabetes, using a semiquantitative reverse transcriptase PCR (RT-PCR) assay. IFN-gamma mRNA levels were significantly higher in the islets obtained from females than those of males, from 10 weeks of age. TNF-beta mRNA was expressed in both females and males between 5 and 15 weeks of age. However, TNF-beta mRNA levels were decreased in males at 20 weeks of age. In contrast, IL-4 mRNA levels were lower in females than in males. These results suggest that islet beta-cell destruction and diabetes in female NOD mice correlates with IFN-gamma and TNF-beta production in the islets, and that male NOD mice may be protected from autoimmune beta-cell destruction by down-regulation of these cytokines. Furthermore, our findings also suggest that insulitis and beta-cell destruction are independently regulated: TNF-beta is more important in forming and maintaining the insulitis, while IFN-gamma has a more important role in beta-cell destruction.
Collapse
Affiliation(s)
- H Hirai
- Department of Pediatrics, Ehime University School of Medicine, Japan
| | | | | | | |
Collapse
|
44
|
Rosen SD. Endothelial ligands for L-selectin: from lymphocyte recirculation to allograft rejection. THE AMERICAN JOURNAL OF PATHOLOGY 1999; 155:1013-20. [PMID: 10514381 PMCID: PMC1867022 DOI: 10.1016/s0002-9440(10)65201-7] [Citation(s) in RCA: 142] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
45
|
Stephens LA, Thomas HE, Ming L, Grell M, Darwiche R, Volodin L, Kay TW. Tumor necrosis factor-alpha-activated cell death pathways in NIT-1 insulinoma cells and primary pancreatic beta cells. Endocrinology 1999; 140:3219-27. [PMID: 10385418 DOI: 10.1210/endo.140.7.6873] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Tumor necrosis factor-alpha (TNFalpha) is a potential mediator of beta cell destruction in insulin-dependent diabetes mellitus. We have studied TNF-responsive pathways leading to apoptosis in beta cells. Primary beta cells express low levels of the type I TNF receptor (TNFR1) but do not express the type 2 receptor (TNFR2). Evidence for TNFR1 expression on beta cells came from flow cytometry using monoclonal antibodies specific for TNFR1 and TNFR2 and from RT-PCR of beta cell RNA. NIT-1 insulinoma cells similarly expressed TNFR1 (at higher levels than primary beta cells) as detected by flow cytometry and radio-binding studies. TNF induced NF-kappaB activation in both primary islet cells and NIT-1 cells. Apoptosis in response to TNFalpha was observed in NIT-1 cells whereas apoptosis of primary beta cells required both TNFalpha and interferon-gamma (IFNgamma). Apoptosis could be prevented in NIT-1 cells by expression of dominant negative Fas-associating protein with death domain (dnFADD). Apoptosis in NIT-1 cells was increased by coincubation with IFNgamma, which also increased caspase 1 expression. These data show that TNF-activated pathways capable of inducing apoptotic cell death are present in beta cells. Caspase activation is the dominant pathway of TNF-induced cell death in NIT-1 cells and may be an important mechanism of beta cell damage in insulin-dependent diabetes mellitus.
Collapse
Affiliation(s)
- L A Stephens
- The Walter and Eliza Hall Institute of Medical Research, Post Office Royal Melbourne Hospital, Parkville, Victoria, Australia
| | | | | | | | | | | | | |
Collapse
|
46
|
Kollias G, Douni E, Kassiotis G, Kontoyiannis D. On the role of tumor necrosis factor and receptors in models of multiorgan failure, rheumatoid arthritis, multiple sclerosis and inflammatory bowel disease. Immunol Rev 1999; 169:175-94. [PMID: 10450517 DOI: 10.1111/j.1600-065x.1999.tb01315.x] [Citation(s) in RCA: 210] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The specific role of the tumor necrosis factor (TNF)/TNF receptor (TNFR) system in disease pathogenesis still remains an unresolved puzzle. Recent studies in transgenic and knockout animals, where the pathogenic influence of genetically perturbed TNF expression has been evaluated, indicate that several pathways of TNF/TNFR action may contribute independently or in concert to initiate, promote or downregulate disease pathogenesis. Evidently, organ-specific inflammatory or autoimmune pathology may ensue due to sustained activation by TNF of innate immune cells and inflammatory responses, which may consequently lead to tissue damage and to organ-specific chronic pathology. However, more cryptic functions of this molecule may be considered to play a significant part in the development of TNF-mediated pathologies. Direct interference of TNF with the differentiation, proliferation or death of specific pathogenic cell targets may be an alternative mechanism for disease initiation or progression. In addition to these activities, there is now considerable evidence to suggest that TNF may also directly promote or downregulate the adaptive immune response. It is therefore evident that no general scenario may adequately describe the role of TNF in disease pathogenesis. In this article, we aim to place these diverse functions of TNF/TNFRs into context with the development of specific pathology in murine models of multiorgan failure, rheumatoid arthritis, multiple sclerosis and inflammatory bowel disease.
Collapse
Affiliation(s)
- G Kollias
- Laboratory of Molecular Genetics, Hellenic Pasteur Institute, Athens, Greece.
| | | | | | | |
Collapse
|
47
|
Crisera CA, Rose MI, Connelly PR, Li M, Colen KL, Longaker MT, Gittes GK. The ontogeny of TGF-beta1, -beta2, -beta3, and TGF-beta receptor-II expression in the pancreas: implications for regulation of growth and differentiation. J Pediatr Surg 1999; 34:689-93; discussion 693-4. [PMID: 10359165 DOI: 10.1016/s0022-3468(99)90357-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND/PURPOSE The transforming growth factor-beta (TGF-beta) cytokines are important regulators of growth and differentiation in multiple mammalian organ systems. Recent studies suggest that they may play a significant role in the regulation of pancreatic organogenesis. The authors proposed to examine the ontogeny of expression of the TGF-beta cytokine isoforms (TGF-beta1, beta2, and beta3), as well as that of the type II TGF-beta receptor (TbetaRII), in the pancreas. We hypothesized that their patterns of expression might help to clarify the manner in which they influence the development of this organ. METHODS Embryos from pregnant CD-1 mice were harvested on gestational days 12.5, 15.5, and 18.5. Microdissection was performed on the embryos to isolate their pancreases. The pancreases were fixed, frozen embedded, and sectioned with a cryostat. Immunohistochemistrywas performed using polyclonal antibodies to TGF-beta1, beta2, and beta3, and TbetaRII. RESULTS The patterns of expression of TGF-beta1, beta2, and beta3 were similar throughout gestation. They were all present, though weakly, early in the development of the pancreas, in the E12.5 epithelial cells. Their expression persisted and became localized to the acinar cells later in gestation. TbetaRII staining was present in both the E12.5 epithelial cells and the surrounding mesenchyme. As the pancreas developed, TbetaRII became strongly expressed in the ductal epithelial cells with only minimal staining in the acinar and endocrine cells. CONCLUSIONS TGF-betas may play a role in regulating pancreatic organogenesis. Our data suggest that they may be required for the normal development of acini. As in other cell systems, TGF-beta1 may act as a suppressor of pancreatic cellular growth and differentiation. The localization of TbetaRII to the mature ductal epithelium may indicate a need for ongoing regulation of growth and differentiation in the pancreatic ducts beyond the fetal period.
Collapse
Affiliation(s)
- C A Crisera
- Laboratory of Developmental Biology and Repair, New York University Medical Center, NY 10016, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Kägi D, Ho A, Odermatt B, Zakarian A, Ohashi PS, Mak TW. TNF Receptor 1-Dependent β Cell Toxicity as an Effector Pathway in Autoimmune Diabetes. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.8.4598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Autoimmune diabetes is characterized by a chronic progressive inflammatory autoimmune reaction that ultimately causes the selective elimination of pancreatic β cells. To address the question of whether the cell death-inducing cytokines TNF and lymphotoxin α are involved in this process, we generated nonobese diabetic (NOD) mice that are deficient for TNF receptor 1 (TNFR1 or TNFRp55). Insulitis developed in these mice similarly to that in normal control NOD mice, but progression to diabetes was completely abrogated. Since this was probably due to the complex immunomodulatory effects of TNF and lymphotoxin α signaled via TNFR1 on lymphohemopoietic cells, adoptive transfer experiments with spleen cells from diabetic NOD mice were conducted. It was found that the absence of TNFR1 in recipients delayed diabetes induced by normal control and precluded diabetes induced by perforin-deficient spleen cells. In a CD8+ T cell-mediated model of diabetes, however, diabetes induced by adoptive transfer of TCR transgenic lymphocytic choriomeningitis virus glycoprotein-specific CD8+ T cells was not delayed by the absence of TNFR1 in recipient mice. Together with the described expression patterns of perforin and TNF in the mononuclear islet infiltrates of NOD mice, these results indicate that two diabetogenic effector mechanisms are delivered by distinct cell populations: CD8+ T cells lyse β cells via perforin-dependent cytotoxicity, whereas CD4+ T cells, macrophages, and dendritic cells contribute to diabetes development via TNFR1-dependent β cell toxicity.
Collapse
Affiliation(s)
- David Kägi
- *Ontario Cancer Institute/Amgen Institute, Toronto, Ontario, Canada; and
| | - Alexandra Ho
- *Ontario Cancer Institute/Amgen Institute, Toronto, Ontario, Canada; and
| | - Bernhard Odermatt
- †Department of Pathology, University of Zürich, Zürich, Switzerland
| | - Arsen Zakarian
- *Ontario Cancer Institute/Amgen Institute, Toronto, Ontario, Canada; and
| | - Pamela S. Ohashi
- *Ontario Cancer Institute/Amgen Institute, Toronto, Ontario, Canada; and
| | - Tak W. Mak
- *Ontario Cancer Institute/Amgen Institute, Toronto, Ontario, Canada; and
| |
Collapse
|
49
|
Pakala SV, Chivetta M, Kelly CB, Katz JD. In autoimmune diabetes the transition from benign to pernicious insulitis requires an islet cell response to tumor necrosis factor alpha. J Exp Med 1999; 189:1053-62. [PMID: 10190896 PMCID: PMC2193009 DOI: 10.1084/jem.189.7.1053] [Citation(s) in RCA: 135] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/1998] [Revised: 01/19/1999] [Indexed: 01/17/2023] Open
Abstract
The islet-infiltrating and disease-causing leukocytes that are a hallmark of insulin-dependent diabetes mellitus produce and respond to a set of cytokine molecules. Of these, interleukin 1beta, tumor necrosis factor (TNF)-alpha, and interferon (IFN)-gamma are perhaps the most important. However, as pleiotropic molecules, they can impact the path leading to beta cell apoptosis and diabetes at multiple points. To understand how these cytokines influence both the formative and effector phases of insulitis, it is critical to determine their effects on the assorted cell types comprising the lesion: the effector T cells, antigen-presenting cells, vascular endothelium, and target islet tissue. Here, we report using nonobese diabetic chimeric mice harboring islets deficient in specific cytokine receptors or cytokine-induced effector molecules to assess how these compartmentalized loss-of-function mutations alter the events leading to diabetes. We found that islets deficient in Fas, IFN-gamma receptor, or inducible nitric oxide synthase had normal diabetes development; however, the specific lack of TNF- alpha receptor 1 (p55) afforded islets a profound protection from disease by altering the ability of islet-reactive, CD4(+) T cells to establish insulitis and subsequently destroy islet beta cells. These results argue that islet cells play a TNF-alpha-dependent role in their own demise.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/pathology
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Autoimmune Diseases/genetics
- Autoimmune Diseases/immunology
- CD4-Positive T-Lymphocytes/immunology
- Chimera
- Diabetes Mellitus, Experimental/immunology
- Diabetes Mellitus, Experimental/therapy
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Disease Progression
- Endothelium, Vascular/pathology
- Gene Targeting
- Islets of Langerhans/immunology
- Islets of Langerhans/physiopathology
- Islets of Langerhans Transplantation
- Mice
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, SCID
- Nephrectomy
- Nitric Oxide Synthase/deficiency
- Nitric Oxide Synthase/genetics
- Nitric Oxide Synthase Type II
- Pancreatitis/genetics
- Pancreatitis/immunology
- Receptors, Interferon/deficiency
- Receptors, Interferon/genetics
- Receptors, Tumor Necrosis Factor/deficiency
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor, Type I
- Receptors, Tumor Necrosis Factor, Type II
- Streptozocin
- Tumor Necrosis Factor-alpha/physiology
- fas Receptor/genetics
- fas Receptor/physiology
- Interferon gamma Receptor
Collapse
Affiliation(s)
- S V Pakala
- Center for Immunology and Department of Pathology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | |
Collapse
|
50
|
Abstract
Insulin-producing B cell tumors (insulinomas) are the most frequent functioning endocrine tumors of the pancreas. Available experimental evidence suggests that the islet B cell is the most likely cell of origin of insulinomas, while the duct endocrine cell should be considered if rearrangement of the pancreatic parenchyma occurs. Data on the genetic background of insulinomas suggest that the B cell tumor development may result from alteration of several genes, including the multiple endocrine neoplasia type 1 (MEN1) gene.
Collapse
|