Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Transplant ; 24(10): 2143-54, 2015.
Article in English | MEDLINE | ID: mdl-26423995

ABSTRACT

Manipulation of regulatory T cell (Treg) migration by islet expression of the chemokine CCL22 prevents diabetes in NOD mice and delays recurrent autoimmunity in syngeneic islet transplants. We sought to determine whether attracting Tregs with CCL22 also prevents islet allograft rejection. Isolated Bl/6 mouse islets were transduced overnight with adenovirus expressing CCL22 (Ad-CCL22) downstream of the CMV promoter. Islets were transplanted under the renal capsule of Balb/c recipients made diabetic by streptozotocin. To assess immunologic tolerance, graft-bearing kidneys from recipients of CCL22-expressing islet grafts were removed, and mice received a second transplant of naive islets from the same donor strain or third-party islets into the contralateral kidney. Adenoviral expression of CCL22 conferred prolonged protection of islet allografts in MHC-mismatched, diabetic recipients, maintaining normoglycemia in 75% of recipients for at least 80 days. Increased frequency of Treg cells was observed in islet grafts transduced with Ad-CCL22 compared with untreated grafts. Normoglycemic recipients of CCL22-expressing islet grafts showed complete absence of antidonor antibodies and no lymphocyte proliferation after exposure to donor splenocytes. After removal of the primary graft at day 80, mice that received a second transplant with untreated islets from the same donor strain did not reject the grafts, suggesting the development of tolerance. Expression of CCL22 recruits Treg cells to transplanted islets, prevents activation of alloreactive T-cells and islet allograft failure and induces alloantigen-specific tolerance. Manipulation of Treg cells by CCL22 in transplanted islets may be a novel therapeutic strategy for diabetes.


Subject(s)
Allografts/immunology , Chemokine CCL22/immunology , Graft Survival/immunology , Immune Tolerance/immunology , Islets of Langerhans Transplantation/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/therapy , Isoantigens/immunology , Mice , Transplantation Tolerance/immunology , Transplantation, Homologous/methods
2.
J Immunol ; 194(7): 3054-64, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25740943

ABSTRACT

Autoimmune destruction of insulin-producing ß cells in type 1 diabetes and islet transplantation involves a variety of immune pathways but is primarily mediated by self-reactive T cells. Chemokines can modulate local immune responses in inflammation and tumors by recruiting immune cells. We have reported that expression of the chemokine CCL22 in pancreatic ß cells in the NOD mouse prevents autoimmune attack by recruiting T regulatory cells (Tregs), protecting mice from diabetes. In this study we show that invariant NKT cells are also recruited to CCL22-expressing islet transplants and are required for CCL22-mediated protection from autoimmunity. Moreover, CCL22 induces an influx of plasmacytoid dendritic cells, which correlates with higher levels of IDO in CCL22-expressing islet grafts. In addition to its chemotactic properties, we found that CCL22 activates Tregs and promotes their ability to induce expression of IDO by dendritic cells. Islet CCL22 expression thus produces a tolerogenic milieu through the interplay of Tregs, invariant NKT cells, and plasmacytoid dendritic cells, which results in suppression of effector T cell responses and protection of ß cells. The immunomodulatory properties of CCL22 could be harnessed for prevention of graft rejection and type 1 diabetes as well as other autoimmune disorders.


Subject(s)
Chemokine CCL22/metabolism , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Animals , Chemokine CCL22/genetics , Chemotaxis/genetics , Chemotaxis/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Diabetes Mellitus, Type 1/genetics , Disease Models, Animal , Female , Gene Expression , Immunomodulation/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans Transplantation , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transduction, Genetic , Transplants/immunology , Transplants/metabolism
3.
J Clin Invest ; 121(8): 3024-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21737880

ABSTRACT

Type 1 diabetes is characterized by destruction of insulin-producing ß cells in the pancreatic islets by effector T cells. Tregs, defined by the markers CD4 and FoxP3, regulate immune responses by suppressing effector T cells and are recruited to sites of action by the chemokine CCL22. Here, we demonstrate that production of CCL22 in islets after intrapancreatic duct injection of double-stranded adeno-associated virus encoding CCL22 recruits endogenous Tregs to the islets and confers long-term protection from autoimmune diabetes in NOD mice. In addition, adenoviral expression of CCL22 in syngeneic islet transplants in diabetic NOD recipients prevented ß cell destruction by autoreactive T cells and thereby delayed recurrence of diabetes. CCL22 expression increased the frequency of Tregs, produced higher levels of TGF-ß in the CD4+ T cell population near islets, and decreased the frequency of circulating autoreactive CD8+ T cells and CD8+ IFN-γ­producing T cells. The protective effect of CCL22 was abrogated by depletion of Tregs with a CD25-specific antibody. Our results indicate that islet expression of CCL22 recruits Tregs and attenuates autoimmune destruction of ß cells. CCL22-mediated recruitment of Tregs to islets may be a novel therapeutic strategy for type 1 diabetes.


Subject(s)
Chemokine CCL22/physiology , Diabetes Mellitus, Type 1/prevention & control , Islets of Langerhans/cytology , T-Lymphocytes, Regulatory/metabolism , Animals , Autoimmune Diseases/metabolism , Autoimmunity , CD4-Positive T-Lymphocytes/metabolism , Chemokine CCL22/genetics , Diabetes Mellitus, Type 1/metabolism , Forkhead Transcription Factors/metabolism , Interleukin-2 Receptor alpha Subunit/biosynthesis , Mice , Mice, Inbred NOD , Mice, SCID , Rats
4.
J Immunol ; 185(4): 2516-24, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20639493

ABSTRACT

IL-1R antagonist (IL-1Ra) is a natural inhibitor of the pleiotropic proinflammatory activities of IL-1. Although several reports described the effects of complete IL-1Ra deficiency, no study has examined the consequences of cell type-specific IL-1Ra inactivation during systemic inflammation. Previous in vitro data demonstrated high IL-1Ra production by hepatocytes and myeloid cells after endotoxin stimulation. In addition, hepatocyte IL-1Ra production is regulated as an acute-phase protein in vitro. In this study, we analyzed the production and functional role of hepatocyte- and myeloid cell-derived IL-1Ra during endotoxin-induced septic shock and acute IL-1beta-induced sterile inflammation. Using conditional IL-1Ra knockout mice, we showed that hepatocytes and myeloid cells are the two major cellular sources of circulating IL-1Ra in response to LPS. Interestingly, IL-1Ra production by myeloid cells, but not hepatocytes, is critical for survival during endotoxemia. Furthermore, we provide the first in vivo evidence demonstrating that IL-1Ra is produced as an acute-phase protein by hepatocytes during IL-1beta-induced inflammation and that hepatocyte-derived IL-1Ra functions as an endogenous negative feedback downregulating the proinflammatory effects of IL-1. Taken together, our observations define distinct roles for two major cellular sources of IL-1Ra in response to different types of systemic inflammatory stimuli in vivo.


Subject(s)
Endotoxemia/blood , Hepatocytes/metabolism , Inflammation/blood , Interleukin 1 Receptor Antagonist Protein/blood , Myeloid Cells/metabolism , Acute-Phase Proteins/genetics , Acute-Phase Proteins/physiology , Animals , Endotoxemia/physiopathology , Enzyme-Linked Immunosorbent Assay , Female , Immunohistochemistry , Inflammation/chemically induced , Inflammation/physiopathology , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin 1 Receptor Antagonist Protein/physiology , Lipopolysaccharides , Liver/metabolism , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Spleen/metabolism , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...