Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Front Immunol ; 13: 1046631, 2022.
Article in English | MEDLINE | ID: mdl-36569931

ABSTRACT

Regulatory T cells (Tregs) normally maintain self-tolerance. Tregs recognize "self" such that when they are not working properly, such as in autoimmunity, the immune system can attack and destroy one's own tissues. Current therapies for autoimmunity rely on relatively ineffective and too often toxic therapies to "treat" the destructive inflammation. Restoring defective endogenous immune regulation (self-tolerance) would represent a paradigm shift in the therapy of these diseases. One recent approach to restore self-tolerance is to use "low dose IL-2" as a therapy to increase the number of circulating Tregs. However, studies to-date have not demonstrated that low-dose IL-2 therapy can restore concomitant Treg function, and phase 2 studies in low dose IL-2 treated patients with autoimmune diseases have failed to demonstrate significant clinical benefit. We hypothesize that the defect in self-tolerance seen in autoimmunity is not due to an insufficient number of available Tregs, but rather, due to defects in second messengers downstream of the IL-2R that normally control Treg function and stability. Previous studies from our lab and others have demonstrated that GRAIL (a ubiquitin E3 ligase) is important in Treg function. GRAIL expression is markedly diminished in Tregs from patients with autoimmune diseases and allergic asthma and is also diminished in Tregs of mice that are considered autoimmune prone. In the relevant pathway in Tregs, GRAIL normally blocks cullin ring ligase activity, which inhibits IL-2R desensitization in Tregs and consequently promotes Treg function. As a result of this defect in GRAIL expression, the Tregs of patients with autoimmune diseases and allergic asthma degrade IL-2R-associated pJAK1 following activation with low dose IL-2, and thus cannot maintain pSTAT5 expression. pSTAT5 controls the transcription of genes required for Treg function. Additionally, the GRAIL-mediated defect may also allow the degradation of the mTOR inhibitor, DEP domain-containing mTOR interacting protein (Deptor). This can lead to IL-2R activation of mTOR and loss of Treg stability in autoimmune patients. Using a monoclonal antibody to the remnant di-glycine tag on ubiquitinated proteins after trypsin digestion, we identified a protein that was ubiquitinated by GRAIL that is important in Treg function, cullin5. Our data demonstrate that GRAIL acts a negative regulator of IL-2R desensitization by ubiquitinating a lysine on cullin5 that must be neddylated to allow cullin5 cullin ring ligase activity. We hypothesize that a neddylation inhibitor in combination with low dose IL-2 activation could be used to substitute for GRAIL and restore Treg function and stability in the Tregs of autoimmune and allergic asthma patients. However, the neddylation activating enzyme inhibitors (NAEi) are toxic when given systemically. By generating a protein drug conjugate (PDC) consisting of a NAEi bound, via cleavable linkers, to a fusion protein of murine IL-2 (to target the drug to Tregs), we were able to use 1000-fold less of the neddylation inhibitor drug than the amount required for therapeutically effective systemic delivery. The PDC was effective in blocking the onset or the progression of disease in several mouse models of autoimmunity (type 1 diabetes, systemic lupus erythematosus, and multiple sclerosis) and a mouse model of allergic asthma in the absence of detectable toxicity. This PDC strategy represents targeted drug delivery at its best where the defect causing the disease was identified, a drug was designed and developed to correct the defect, and the drug was targeted and delivered only to cells that needed it, maximizing safety and efficacy.


Subject(s)
Autoimmune Diseases , T-Lymphocytes, Regulatory , Mice , Animals , Interleukin-2/metabolism , Cullin Proteins/metabolism , Receptors, Interleukin-2 , Autoimmune Diseases/metabolism , TOR Serine-Threonine Kinases/metabolism
2.
Diabetes ; 71(7): 1490-1507, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35499603

ABSTRACT

Multiple pathways contribute to the pathophysiological development of type 1 diabetes (T1D); however, the exact mechanisms involved are unclear. We performed differential gene expression analysis in pancreatic islets of NOD mice versus age-matched congenic NOD.B10 controls to identify genes that may contribute to disease pathogenesis. Novel genes related to extracellular matrix development and glucagon and insulin signaling/secretion were changed in NOD mice during early inflammation. During "respective" insulitis, the expression of genes encoding multiple chemosensory olfactory receptors were upregulated, and during "destructive" insulitis, the expression of genes involved in antimicrobial defense and iron homeostasis were downregulated. Islet inflammation reduced the expression of Hamp that encodes hepcidin. Hepcidin is expressed in ß-cells and serves as the key regulator of iron homeostasis. We showed that Hamp and hepcidin levels were lower, while iron levels were higher in the pancreas of 12-week-old NOD versus NOD.B10 mice, suggesting that a loss of iron homeostasis may occur in the islets during the onset of "destructive" insulitis. Interestingly, we showed that the severity of NOD disease correlates with dietary iron intake. NOD mice maintained on low-iron diets had a lower incidence of hyperglycemia, while those maintained on high-iron diets had an earlier onset and higher incidence of disease, suggesting that high iron exposure combined with a loss of pancreatic iron homeostasis may exacerbate NOD disease. This mechanism may explain the link seen between high iron exposure and the increased risk for T1D in humans.


Subject(s)
Diabetes Mellitus, Type 1 , Islets of Langerhans , Animals , Diabetes Mellitus, Type 1/metabolism , Hepcidins/genetics , Hepcidins/metabolism , Homeostasis/genetics , Inflammation/metabolism , Iron/metabolism , Islets of Langerhans/metabolism , Mice , Mice, Inbred NOD , Pancreas/metabolism
3.
Biomedicines ; 8(3)2020 Mar 04.
Article in English | MEDLINE | ID: mdl-32143316

ABSTRACT

The antigen-specific apoptotic DNA immunotherapeutic, ADi-100, is designed to suppress type 1 diabetes and consists of two DNA plasmids encoding genetic sequences of the apoptosis-inducing molecule, BAX, and the secreted form of the autoantigen, glutamic acid decarboxylase 65, that is CpG hyper-methylated to avoid inflammatory signaling (msGAD55). Upon a four-day treatment with ADi-100 of young female non-obese diabetic (NOD) mice, the frequency of various tolerogenic dendritic cell populations increased in draining lymph nodes; these cells lost the capacity to stimulate glutamic acid decarboxylase (GAD)-specific CD4+ T lymphocytes and were associated with the previously demonstrated enhancement of GAD-specific regulatory T cells. The efficacy of two ADi-100 formulations containing different proportions of BAX and msGAD55, 1:4 (10/40 µg) and 1:2 (17/33 µg), was evaluated in mildly hyperglycemic pre-diabetic NOD female mice. Both formulations suppressed the incidence of diabetes by 80% in an antigen-specific manner, while all untreated mice developed diabetes. However, treatment of pre-diabetic mice with significantly higher hyperglycemia, denoting progressive disease, showed that ADi-100 1:2 strongly suppressed diabetes incidence by 80% whereas the ADi-100 1:4 was less effective (50%). As an antigen-specific monotherapy, ADi-100 is highly efficacious in reversing elevated hyperglycemia to prevent diabetes, in which increasing apoptosis-inducing BAX content is a promising immune tolerance feature.

4.
Front Endocrinol (Lausanne) ; 11: 609271, 2020.
Article in English | MEDLINE | ID: mdl-33424774

ABSTRACT

Type 1 Diabetes (T1D) occurs as a result of the autoimmune destruction of pancreatic ß-cells by self-reactive T cells. The etiology of this disease is complex and difficult to study due to a lack of disease-relevant tissues from pre-diabetic individuals. In this study, we performed gene expression analysis on human pancreas tissues obtained from the Network of Pancreatic Organ Donors with Diabetes (nPOD), and showed that 155 genes were differentially expressed by ≥2-fold in the pancreata of autoantibody-positive (AA+) at-risk individuals compared to healthy controls. Only 48 of these genes remained changed by ≥2-fold in the pancreata of established T1D patients. Pathway analysis of these genes showed a significant association with various immune pathways. We were able to validate the differential expression of eight disease-relevant genes by QPCR analysis: A significant upregulation of CADM2, and downregulation of TRPM5, CRH, PDK4, ANGPL4, CLEC4D, RSG16, and FCGR2B was confirmed in the pancreata of AA+ individuals versus controls. Studies have already implicated FCGR2B in the pathogenesis of disease in non-obese diabetic (NOD) mice. Here we showed that CADM2, TRPM5, PDK4, and ANGPL4 were similarly changed in the pancreata of pre-diabetic 12-week-old NOD mice compared to NOD.B10 controls, suggesting a possible role for these genes in the pathogenesis of both T1D and NOD disease. The loss of the leukocyte-specific gene, FCGR2B, in the pancreata of AA+ individuals, is particularly interesting, as it may serve as a potential whole blood biomarker of disease progression. To test this, we quantified FCGR2B expression in peripheral blood samples of T1D patients, and AA+ and AA- first-degree relatives of T1D patients enrolled in the TrialNet Pathway to Prevention study. We showed that FCGR2B was significantly reduced in the peripheral blood of AA+ individuals compared to AA- controls. Together, these findings demonstrate that gene expression analysis of pancreatic tissue and peripheral blood samples can be used to identify disease-relevant genes and pathways and potential biomarkers of disease progression in T1D.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Gene Expression Profiling/methods , Pancreas/chemistry , Prediabetic State/genetics , Adolescent , Adult , Animals , Autoantibodies/genetics , Autoantibodies/immunology , Biomarkers/analysis , Child , Diabetes Mellitus, Type 1/pathology , Disease Progression , Female , Gene Expression Regulation , Humans , Infant , Male , Mice , Mice, Inbred NOD , Microarray Analysis , Middle Aged , Prognosis , RNA/genetics , Receptors, IgG/genetics , Signal Transduction/genetics , Young Adult
5.
Crit Rev Immunol ; 40(3): 185-193, 2020.
Article in English | MEDLINE | ID: mdl-33389883

ABSTRACT

We identified a druggable defect in IL-2 receptor (IL-2R) signaling by comparing the response of regulatory T cells (Tregs) of autoimmune disease patients to that of healthy controls. This defect was in the inhibition of Treg desensitization and was shared across various autoimmune diseases. Low-dose IL-2 stimulation results in maintained pSTAT5 expression for > 4 h, allowing the Treg transcriptome for "function" to be transcribed. Tregs of autoimmune Tregs of autoimmune disease patients more rapidly terminate IL-2R signaling through STAT5. Prolonged pSTAT5 expression following IL-2R activation is mediated by blocking proteasomal degradation of pJAKl, which is associated with the IL-2RP chain. In Tregs of controls, this is accomplished by inhibiting a requisite-activating post-translational modification (neddylation) of the SOCS3/Cul5 cullin ring ligase (CRL), which normally ubiquitinates pJAKl. Many receptor-associated tyrosine kinases are desensitized by a CRL. Tregs uniquely constitutively express an E3 ligase known as the gene related to anergy in lymphocytes (GRAIL), which ubiquinates the exact lysine on the Cul5 protein that needs to be neddylated as a condition for the activation and consequent ubiquitination of pJAKl. There is a defect in this GRAIL-associated pathway of competitive inhibition of neddylation in the Tregs of autoimmune disease patients. This defect can be mitigated by the application of a small-molecule drug known as a neddylation activating enzyme inhibitor (NAEi). Low-dose IL-2 and an NAEi as a protein-drug conjugate was found to be much more effective than simply using low-dose IL-2 or a combination of low-dose IL-2 and an NAEi systemically in treating animal models of autoimmune diseases.


Subject(s)
Autoimmune Diseases/drug therapy , Immunosuppressive Agents/pharmacology , Receptors, Interleukin-2/metabolism , T-Lymphocytes, Regulatory/drug effects , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Disease Models, Animal , Drug Evaluation, Preclinical , Gene Expression Profiling , Humans , Immunosuppressive Agents/therapeutic use , Janus Kinase 1/metabolism , Mice , Phosphorylation/drug effects , Phosphorylation/immunology , Proteolysis , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects , Ubiquitination/immunology
6.
BMC Genomics ; 18(1): 636, 2017 Aug 18.
Article in English | MEDLINE | ID: mdl-28821222

ABSTRACT

BACKGROUND: The natural history of type 1 diabetes (T1D) is challenging to investigate, especially as pre-diabetic individuals are difficult to identify. Numerous T1D consortia have been established to collect whole blood for gene expression analysis from individuals with or at risk to develop T1D. However, with no universally accepted protocol for their collection, differences in sample processing may lead to variances in the results. Here, we examined whether the choice of blood collection tube and RNA extraction kit leads to differences in the expression of genes that are changed during the progression of T1D, and if these differences could be minimized by measuring gene expression directly from the lysate of whole blood. RESULTS: Microarray analysis showed that the expression of 901 genes is highly influenced by sample processing using the PAXgene versus the Tempus system. These included a significant number of lymphocyte-specific genes and genes whose expression has been reported to differ in the peripheral blood of at-risk and T1D patients compared to controls. We showed that artificial changes in gene expression occur when control and T1D samples were processed differently. The sample processing-dependent differences in gene expression were largely due to loss of transcripts during the RNA extraction step using the PAXgene system. The majority of differences were not observed when gene expression was measured in whole blood lysates prepared from blood collected in PAXgene and Tempus tubes. CONCLUSION: We showed that the gene expression profile of samples processed using the Tempus system is more accurate than that of samples processed using the PAXgene system. Variation in sample processing can result in misleading changes in gene expression. However, these differences can be minimized by measuring gene expression directly in whole blood lysates.


Subject(s)
Blood Specimen Collection/methods , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/genetics , Gene Expression Profiling/methods , Leukocytes, Mononuclear/metabolism , Adolescent , Adult , Aged , Female , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Young Adult
7.
Diabetes ; 65(5): 1341-9, 2016 May.
Article in English | MEDLINE | ID: mdl-26983959

ABSTRACT

Type 1 diabetes (T1D) is caused by autoreactive T cells that recognize pancreatic islet antigens and destroy insulin-producing ß-cells. This attack results from a breakdown in tolerance for self-antigens, which is controlled by ectopic antigen expression in the thymus and pancreatic lymph nodes (PLNs). The autoantigens known to be involved include a set of islet proteins, such as insulin, GAD65, IA-2, and ZnT8. In an attempt to identify additional antigenic proteins, we performed an expression-based genome-wide association study using microarray data from 118 arrays of the thymus and PLNs of T1D mice. We ranked all 16,089 protein-coding genes by the likelihood of finding repeated differential expression and the degree of tissue specificity for pancreatic islets. The top autoantigen candidate was vitamin D-binding protein (VDBP). T-cell proliferation assays showed stronger T-cell reactivity to VDBP compared with control stimulations. Higher levels and frequencies of serum anti-VDBP autoantibodies (VDBP-Abs) were identified in patients with T1D (n = 331) than in healthy control subjects (n = 77). Serum vitamin D levels were negatively correlated with VDBP-Ab levels in patients in whom T1D developed during the winter. Immunohistochemical localization revealed that VDBP was specifically expressed in α-cells of pancreatic islets. We propose that VDBP could be an autoantigen in T1D.


Subject(s)
Autoantigens/metabolism , Autoimmune Diseases/metabolism , Autoimmunity , Diabetes Mellitus, Type 1/metabolism , Gene Expression Regulation, Developmental , Glucagon-Secreting Cells/metabolism , Vitamin D-Binding Protein/metabolism , Adolescent , Animals , Autoantigens/genetics , Autoimmune Diseases/blood , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Case-Control Studies , Cells, Cultured , Child , Child, Preschool , Colorado , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Female , Genome-Wide Association Study , Glucagon-Secreting Cells/immunology , Glucagon-Secreting Cells/pathology , Humans , Male , Mice, Inbred NOD , Organ Specificity , Seasons , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Spleen/pathology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Vitamin D/analogs & derivatives , Vitamin D/blood , Vitamin D-Binding Protein/genetics
8.
J Infect Dis ; 213(3): 456-64, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26150546

ABSTRACT

T cell suppression in sepsis is a well-known phenomenon; however, the underlying mechanisms are not fully understood. Previous studies have shown that T cell stimulation up-regulates mitochondrial adenosine triphosphate (ATP) production to fuel purinergic signaling mechanisms necessary for adequate T cell responses. Here we show that basal mitochondrial ATP production, ATP release, and stimulation of P2X1 receptors represent a standby purinergic signaling mechanism that is necessary for antigen recognition. Inhibition of this process impairs T cell vigilance and the ability of T cells to trigger T cell activation, up-regulate mitochondrial ATP production, and stimulate P2X4 and P2X7 receptors that elicit interleukin 2 production and T cell proliferation. T cells of patients with sepsis lack this standby purinergic signaling system owing to defects in mitochondrial function, ATP release, and calcium signaling. These defects impair antigen recognition and T cell function and are correlated with sepsis severity. Pharmacological targeting of these defects may improve T cell function and reduce the risk of sepsis.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Calcium Signaling/physiology , Mitochondria/physiology , Purines/metabolism , Receptors, Purinergic/physiology , Sepsis/immunology , Adolescent , Adult , Humans , Jurkat Cells , Suramin , Young Adult
9.
Diabetes ; 64(2): 604-17, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25187368

ABSTRACT

Peripheral tolerance is partially controlled by the expression of peripheral tissue antigens (PTAs) in lymph node stromal cells (LNSCs). We previously identified a transcriptional regulator, deformed epidermal autoregulatory factor 1 (Deaf1), that can regulate PTA expression in LNSCs of the pancreatic lymph nodes (PLNs). During the pathogenesis of type 1 diabetes (T1D), Deaf1 is spliced to form the dominant-negative isoform Deaf1-Var1. Here we show that Deaf1-Var1 expression correlates with the severity of disease in NOD mice and is reduced in the PLNs of mice that do not develop hyperglycemia. Inflammation and hyperglycemia independently drive Deaf1 splicing through activation of the splicing factors Srsf10 and Ptbp2, respectively. Inflammation induced by injection of activated splenocytes increased Deaf1-Var1 and Srsf10, but not Ptbp2, in the PLNs of NOD.SCID mice. Hyperglycemia induced by treatment with the insulin receptor agonist S961 increased Deaf1-Var1 and Ptbp2, but not Srsf10, in the PLNs of NOD.B10 and NOD mice. Overexpression of PTBP2 and/or SRSF10 also increased human DEAF1-VAR1 and reduced PTA expression in HEK293T cells. These data suggest that during the progression of T1D, inflammation and hyperglycemia mediate the splicing of DEAF1 and loss of PTA expression in LNSCs by regulating the expression of SRSF10 and PTBP2.


Subject(s)
Alternative Splicing , Diabetes Mellitus, Type 1/metabolism , Hyperglycemia/metabolism , Inflammation/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Aging , Animals , Blood Glucose , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA-Binding Proteins , Diabetes Mellitus, Type 1/genetics , Female , HEK293 Cells , Humans , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Lymph Nodes/physiology , Mice , Mice, Inbred NOD , Mice, SCID , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Pancreas/physiology , Pancreatic Elastase/genetics , Pancreatic Elastase/metabolism , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Serine-Arginine Splicing Factors , Spleen/cytology , Transcription Factors/genetics
10.
Curr Diab Rep ; 14(9): 525, 2014.
Article in English | MEDLINE | ID: mdl-25030265

ABSTRACT

Type 1 diabetes (T1D) may result from a breakdown in peripheral tolerance that is partially controlled by the ectopic expression of peripheral tissue antigens (PTAs) in lymph nodes. Various subsets of lymph node stromal cells and certain hematopoietic cells play a role in maintaining T cell tolerance. These specialized cells have been shown to endogenously transcribe, process, and present a range of PTAs to naive T cells and mediate the clonal deletion or inactivation of autoreactive cells. During the progression of T1D, inflammation leads to reduced PTA expression in the pancreatic lymph nodes and the production of novel islet antigens that T cells are not tolerized against. These events allow for the escape and activation of autoreactive T cells and may contribute to the pathogenesis of T1D. In this review, we discuss recent findings in this area and propose possible therapies that may help reestablish self-tolerance during T1D.


Subject(s)
Autoantigens/immunology , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/immunology , Immune System/immunology , Self Tolerance/immunology , Alternative Splicing/genetics , Animals , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/therapy , Humans , Lymph Nodes/immunology , Lymph Nodes/pathology
11.
Immunol Res ; 58(2-3): 340-50, 2014 May.
Article in English | MEDLINE | ID: mdl-24682832

ABSTRACT

Type 1 diabetes (T1D) is a complex polygenic disease that is triggered by various environmental factors in genetically susceptible individuals. The emphasis placed on genome-wide association studies to explain the genetics of T1D has failed to advance our understanding of T1D pathogenesis or identify biomarkers of disease progression or therapeutic targets. Using the nonobese diabetic (NOD) mouse model of T1D and the non-disease prone congenic NOD.B10 mice, our laboratory demonstrated striking tissue-specific and age-dependent changes in gene expression during disease progression. We established a "roadmap" of differential gene expression and used this to identify candidate genes in mice (and human orthologs) that play a role in disease pathology. Here, we describe two genes, Deformed epidermal autoregulatory factor 1 (Deaf1) and Adenosine A1 receptor (Adora1), that are differentially expressed and alternatively spliced in the pancreatic lymph nodes or islets of NOD mice and T1D patients to form dominant-negative non-functional isoforms. Loss of Deaf1 function leads to reduced peripheral tissue antigen expression in lymph node stromal cells and may contribute to a breakdown in peripheral tolerance, while reduced Adora1 function results in an early intrinsic alpha cell defect that may explain the hyperglucagonemia and resulting beta cell stress observed prior to the onset of diabetes. Remarkably, both genes were also alternatively spliced in the same tissues of auto-antibody positive prediabetic patients, and these splicing events resulted in similar downstream effects as those seen in NOD mice. These findings demonstrate the value of gene expression profiling in studying disease pathogenesis in T1D.


Subject(s)
Diabetes Mellitus, Type 1/etiology , Gene Expression Profiling , Gene Expression Regulation , Alternative Splicing , Animals , Antigen Presentation/immunology , Antigens/genetics , Antigens/immunology , DNA-Binding Proteins , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/therapy , Disease Models, Animal , Humans , Islets of Langerhans/cytology , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Mice , Mice, Inbred NOD , Nuclear Proteins/genetics , Organ Specificity/genetics , Receptor, Adenosine A1/metabolism , Transcription Factors , Translational Research, Biomedical
12.
Diabetes ; 62(12): 4208-19, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24264405

ABSTRACT

Prediabetic NOD mice exhibit hyperglucagonemia, possibly due to an intrinsic α-cell defect. Here, we show that the expression of a potential glucagon inhibitor, the adenosine A1 receptor (Adora1), is gradually diminished in α-cells of NOD mice, autoantibody-positive (AA(+)) and overtly type 1 diabetic (T1D) patients during the progression of disease. We demonstrated that islet inflammation was associated with loss of Adora1 expression through the alternative splicing of Adora1. Expression of the spliced variant (Adora1-Var) was upregulated in the pancreas of 12-week-old NOD versus age-matched NOD.B10 (non-diabetes-susceptible) control mice and was detected in the pancreas of AA(+) patients but not in control subjects or overtly diabetic patients, suggesting that inflammation drives the splicing of Adora1. We subsequently demonstrated that Adora1-Var expression was upregulated in the islets of NOD.B10 mice after exposure to inflammatory cytokines and in the pancreas of NOD.SCID mice after adoptive transfer of activated autologous splenocytes. Adora1-Var encodes a dominant-negative N-terminal truncated isoform of Adora1. The splicing of Adora1 and loss of Adora1 expression on α-cells may explain the hyperglucagonemia observed in prediabetic NOD mice and may contribute to the pathogenesis of human T1D and NOD disease.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Glucagon-Secreting Cells/metabolism , Prediabetic State/metabolism , Receptor, Adenosine A1/metabolism , Adolescent , Adult , Animals , Child , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Female , Humans , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred NOD , Middle Aged , Prediabetic State/genetics , Prediabetic State/pathology , Receptor, Adenosine A1/genetics , Spleen/metabolism , Spleen/pathology
13.
J Mol Cell Biol ; 5(2): 99-110, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22923498

ABSTRACT

The transcriptional regulator deformed epidermal autoregulatory factor 1 (DEAF1) has been suggested to play a role in maintaining peripheral tolerance by controlling the transcription of peripheral tissue antigen genes in lymph node stromal cells (LNSCs). Here, we demonstrate that DEAF1 also regulates the translation of genes in LNSCs by controlling the transcription of the poorly characterized eukaryotic translation initiation factor 4 gamma 3 (Eif4g3) that encodes eIF4GII. Eif4g3 gene expression was reduced in the pancreatic lymph nodes of Deaf1-KO mice, non-obese diabetic mice, and type 1 diabetes patients, where functional Deaf1 is absent or diminished. Silencing of Deaf1 reduced Eif4g3 expression, but increased the expression of Caspase 3, a serine protease that degrades eIF4GII. Polysome profiling showed that reduced Eif4g3 expression in LNSCs resulted in the diminished translation of various genes, including Anpep, the gene for aminopeptidase N, an enzyme involved in fine-tuning antigen presentation on major histocompatibility complex (MHC) class II. Together these findings suggest that reduced DEAF1 function, and subsequent loss of Eif4g3 transcription may affect peripheral tissue antigen (PTA) expression in LNSCs and contribute to the pathology of T1D.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Eukaryotic Initiation Factor-4G/biosynthesis , Lymph Nodes/metabolism , Pancreas/metabolism , Protein Biosynthesis , Transcription Factors/metabolism , Animals , Caspase 3/biosynthesis , Caspase 3/genetics , Caspase 3/immunology , DNA-Binding Proteins , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Eukaryotic Initiation Factor-4G/genetics , Eukaryotic Initiation Factor-4G/immunology , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Immune Tolerance/genetics , Lymph Nodes/immunology , Lymph Nodes/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Knockout , Pancreas/immunology , Pancreas/pathology , Stromal Cells/immunology , Stromal Cells/metabolism , Stromal Cells/pathology , Transcription Factors/genetics , Transcription Factors/immunology
14.
Regul Pept ; 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21256874

ABSTRACT

This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.

15.
J Pharmacol Exp Ther ; 336(1): 77-86, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20876230

ABSTRACT

Ghrelin, a potent orexigenic hormone released from the stomach, is important in regulating energy metabolism. Abnormal ghrelin levels are associated with eating disorders and metabolic diseases. However, factors involved in the regulation of ghrelin release remain unclear. Here, we examined the involvement of adenosine signaling in the control of ghrelin release from the perfused mouse stomach. Adenosine stimulated ghrelin release concentration-dependently, and the A(2A) receptor-selective antagonists 4-(2-[7-amino-2-(2-furyl)[1,2,4]triazolo[2,3-a][1,3,5]triazin-5-ylamino]ethyl)phenol (ZM 241385) and 2-(2-furanyl)-7-(2-phenylethyl)-7H-pyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidin-5-amine (SCH 58261) abolished the increased release. The A(2A) receptor-selective agonist 2-p-(2-carboxyethyl)phenethylamino-5-N-ethylcarboxamidoadenosine hydrochloride (CGS 21680) augmented ghrelin release concentration-dependently, whereas the A(1) receptor-selective agonist 2-chloro-N(6)-cyclopentyladenosine inhibited ghrelin release. In A(2A) receptor knockout mice, adenosine inhibited ghrelin release, and the A(1) receptor-selective antagonist 8-cyclopentyl-1,3-dipropylxanthine blocked this inhibition. The adenosine deaminase inhibitor erythro-9-(2-hydroxy-3-nonyl)adenine hydrochloride increased ghrelin release in wild-type and A(1) receptor knockout mice but not in A(2A) receptor knockout mice. Colocalization of ghrelin immunoreactivity with A(1) and A(2A) receptor immunoreactivities in the gastric nerve fibers were observed. Colocalization was also detected for ghrelin and A(1) receptor immunoreactivities in the gastric mucosa. Blockade of neural activities with tetrodotoxin abolished the stimulatory effect of adenosine on ghrelin release. In conclusion, adenosine exerts predominantly a tonic A(2A) receptor-mediated stimulatory action on gastric ghrelin release, whereas an A(1) receptor-mediated inhibitory action is also apparent when the tonic excitatory effect was removed.


Subject(s)
Adenosine/physiology , Gastric Mucosa/metabolism , Ghrelin/metabolism , Receptor, Adenosine A1/physiology , Receptor, Adenosine A2A/physiology , Signal Transduction/physiology , Animals , Dose-Response Relationship, Drug , Gastric Mucosa/drug effects , Male , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Perfusion , Receptor, Adenosine A1/deficiency , Receptor, Adenosine A2A/deficiency , Signal Transduction/drug effects
16.
J Leukoc Biol ; 88(6): 1181-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20884646

ABSTRACT

Hypertonic saline (HS) resuscitation increases T cell function and inhibits posttraumatic T cell anergy, which can reduce immunosuppression and sepsis in trauma patients. We have previously shown that HS induces the release of cellular ATP and enhances T cell function. However, the mechanism by which HS induces ATP release and the subsequent regulation of T cell function by ATP remain poorly understood. In the present study, we show that inhibition of the gap junction hemichannel pannexin-1 (Panx1) blocks ATP release in response to HS, and HS exposure triggers significant changes in the expression of all P2X-type ATP receptors in Jurkat T cells. Blocking or silencing of Panx1 or of P2X1, P2X4, or P2X7 receptors blunts HS-induced p38 MAPK activation and the stimulatory effects of HS on TCR/CD28-induced IL-2 gene transcription. Moreover, treatment with HS or agonists of P2X receptors overcomes T cell suppression induced by the anti-inflammatory cytokine IL-10. These findings indicate that Panx1 hemichannels facilitate ATP release in response to hypertonic stress and that P2X1, P2X4, and P2X7 receptor activation enhances T cell function. We conclude that HS and P2 receptor agonists promote T cell function and thus, could be used to improve T cell function in trauma patients.


Subject(s)
Connexins/physiology , Nerve Tissue Proteins/physiology , Receptors, Purinergic P2X/physiology , Saline Solution, Hypertonic/pharmacology , Stress, Physiological/immunology , T-Lymphocytes/physiology , Adenosine Triphosphate/metabolism , Gadolinium/pharmacology , Humans , Interleukin-10/physiology , Interleukin-2/genetics , Jurkat Cells , Receptors, Purinergic P2X1/physiology , Receptors, Purinergic P2X4/physiology , Receptors, Purinergic P2X7/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Blood ; 116(18): 3475-84, 2010 Nov 04.
Article in English | MEDLINE | ID: mdl-20660288

ABSTRACT

Engagement of T cells with antigen-presenting cells requires T-cell receptor (TCR) stimulation at the immune synapse. We previously reported that TCR stimulation induces the release of cellular adenosine-5'-triphosphate (ATP) that regulates T-cell activation. Here we tested the roles of pannexin-1 hemichannels, which have been implicated in ATP release, and of various P2X receptors, which serve as ATP-gated Ca(2+) channels, in events that control T-cell activation. TCR stimulation results in the translocation of P2X1 and P2X4 receptors and pannexin-1 hemichannels to the immune synapse, while P2X7 receptors remain uniformly distributed on the cell surface. Removal of extracellular ATP or inhibition, mutation, or silencing of P2X1 and P2X4 receptors inhibits Ca(2+) entry, nuclear factors of activated T cells (NFAT) activation, and induction of interleukin-2 synthesis. Inhibition of pannexin-1 hemichannels suppresses TCR-induced ATP release, Ca(2+) entry, and T-cell activation. We conclude that pannexin-1 hemichannels and P2X1 and P2X4 receptors facilitate ATP release and autocrine feedback mechanisms that control Ca(2+) entry and T-cell activation at the immune synapse.


Subject(s)
Adenosine Triphosphate/immunology , Connexins/immunology , Immunological Synapses/immunology , Nerve Tissue Proteins/immunology , Receptors, Purinergic P2X1/immunology , Receptors, Purinergic P2X4/immunology , T-Lymphocytes/immunology , Calcium/immunology , Calcium Channels/genetics , Connexins/metabolism , Gene Expression , Humans , Immunological Synapses/ultrastructure , Interleukin-2/genetics , Interleukin-2/immunology , Jurkat Cells , Lymphocyte Activation , Membrane Proteins/genetics , NFATC Transcription Factors/immunology , Neoplasm Proteins/genetics , Nerve Tissue Proteins/metabolism , ORAI1 Protein , Protein Transport , Receptors, Purinergic P2X1/genetics , Receptors, Purinergic P2X4/genetics , Receptors, Purinergic P2X5/genetics , Receptors, Purinergic P2X5/immunology , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/immunology , Stromal Interaction Molecule 1 , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , T-Lymphocytes/ultrastructure
18.
Nat Immunol ; 10(9): 1026-33, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19668219

ABSTRACT

Type 1 diabetes may result from a breakdown in peripheral tolerance that is partially controlled by the expression of peripheral tissue antigens (PTAs) in lymph nodes. Here we show that the transcriptional regulator Deaf1 controls the expression of genes encoding PTAs in the pancreatic lymph nodes (PLNs). The expression of canonical Deaf1 was lower, whereas that of an alternatively spliced variant was higher, during the onset of destructive insulitis in the PLNs of nonobese diabetic (NOD) mice. We identified an equivalent variant Deaf1 isoform in the PLNs of patients with type 1 diabetes. Both the NOD mouse and human Deaf1 variant isoforms suppressed PTA expression by inhibiting the transcriptional activity of canonical Deaf1. Lower PTA expression resulting from the alternative splicing of DEAF1 may contribute to the pathogenesis of type 1 diabetes.


Subject(s)
Antigens/genetics , Diabetes Mellitus, Type 1/immunology , Lymph Nodes/immunology , Nuclear Proteins/physiology , Pancreas/immunology , Alternative Splicing , Animals , Base Sequence , DNA-Binding Proteins , Female , Gene Expression Regulation , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Molecular Sequence Data , Protein Isoforms , Transcription Factors
19.
FASEB J ; 23(6): 1685-93, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19211924

ABSTRACT

T-cell activation requires the influx of extracellular calcium, although mechanistic details regarding such activation are not fully defined. Here, we show that P2X(7) receptors play a key role in calcium influx and downstream signaling events associated with the activation of T cells. By real-time PCR and immunohistochemistry, we find that Jurkat T cells and human CD4(+) T cells express abundant P2X(7) receptors. We show, using a novel fluorescent microscopy technique, that T-cell receptor (TCR) stimulation triggers the rapid release of ATP (<100 microM). This release of ATP is required for TCR-mediated calcium influx, NFAT activation, and interleukin-2 (IL-2) production. TCR activation up-regulates P2X(7) receptor gene expression. Removal of extracellular ATP by apyrase or alkaline phosphatase treatment, inhibition of ATP release with the maxi-anion channel blocker gadolinium chloride, or siRNA silencing of P2X(7) receptors blocks calcium entry and inhibits T-cell activation. Moreover, lymphocyte activation is impaired in C57BL/6 mice that express poorly functional P2X(7) receptors, compared to control BALB/c mice, which express fully functional P2X(7) receptors. We conclude that ATP release and autocrine, positive feedback through P2X(7) receptors is required for the effective activation of T cells.


Subject(s)
Adenosine Triphosphate/metabolism , Autocrine Communication/physiology , CD4-Positive T-Lymphocytes/metabolism , Calcium Signaling/physiology , Lymphocyte Activation , Receptors, Purinergic P2/metabolism , Animals , CD4-Positive T-Lymphocytes/cytology , Calcium/metabolism , Humans , Interleukin-2/genetics , Interleukin-2/metabolism , Jurkat Cells , Mice , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X7
20.
Shock ; 30(2): 173-7, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18091570

ABSTRACT

We have recently shown that A3 adenosine receptors and P2Y2 purinergic receptors play an important role in neutrophil chemotaxis. Chemotaxis of neutrophils to sites of infections is critical for immune defense. However, excessive accumulation of neutrophils in the lungs can cause acute lung tissue damage. Here we assessed the role of A3 and P2Y2 receptors in neutrophil sequestration to the lungs in a mouse model of sepsis. Sepsis was induced by cecal ligation and puncture (CLP) using adult male C57BL/6J mice (wild type [WT]), homozygous A3 receptor knockout (A3KO) mice, and P2Y2 receptor knockout (P2Y2KO) mice. Animals were killed 2, 4, 6, or 8 h after CLP, and peritoneal lavage fluid and blood were collected. Lungs were removed, and neutrophil infiltration was evaluated using elastase as a marker. Leukocyte and bacterial counts in peritoneal lavage fluid and blood samples were determined. Survival after sepsis was determined in a separate group. Leukocyte counts in the peritoneum were lower in A3KO and P2Y2KO mice than in WT mice. Conversely, initial leukocyte counts in the peripheral blood were higher in KO mice than in WT mice. Neutrophil sequestration to the lungs reached a maximum 2 h after CLP and remained significantly higher in WT mice compared with A3KO and P2Y2KO mice (P < 0.001). Survival after 24 h was significantly lower in WT mice (37.5%) than in A3KO or P2Y2KO mice (82.5%; P < 0.05). These data suggest that A3 and P2Y2 receptors are involved in the influx of neutrophils into the lungs after sepsis. Thus, pharmaceutical approaches that target these receptors might be useful to control acute lung tissue injury in sepsis.


Subject(s)
Disease Models, Animal , Lung/pathology , Neutrophil Infiltration/physiology , Neutrophils/metabolism , Neutrophils/pathology , Receptor, Adenosine A3/physiology , Receptors, Purinergic P2/physiology , Sepsis/pathology , Animals , Cecum , Ligation , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Punctures , Receptor, Adenosine A3/deficiency , Receptor, Adenosine A3/genetics , Receptors, Purinergic P2/deficiency , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2Y2 , Sepsis/metabolism , Sepsis/mortality
SELECTION OF CITATIONS
SEARCH DETAIL
...