Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Diabet Med ; 37(12): 2160-2168, 2020 12.
Article in English | MEDLINE | ID: mdl-32634859

ABSTRACT

AIMS: Misclassification of diabetes is common due to an overlap in the clinical features of type 1 and type 2 diabetes. Combined diagnostic models incorporating clinical and biomarker information have recently been developed that can aid classification, but they have not been validated using pancreatic pathology. We evaluated a clinical diagnostic model against histologically defined type 1 diabetes. METHODS: We classified cases from the Network for Pancreatic Organ donors with Diabetes (nPOD) biobank as type 1 (n = 111) or non-type 1 (n = 42) diabetes using histopathology. Type 1 diabetes was defined by lobular loss of insulin-containing islets along with multiple insulin-deficient islets. We assessed the discriminative performance of previously described type 1 diabetes diagnostic models, based on clinical features (age at diagnosis, BMI) and biomarker data [autoantibodies, type 1 diabetes genetic risk score (T1D-GRS)], and singular features for identifying type 1 diabetes by the area under the curve of the receiver operator characteristic (AUC-ROC). RESULTS: Diagnostic models validated well against histologically defined type 1 diabetes. The model combining clinical features, islet autoantibodies and T1D-GRS was strongly discriminative of type 1 diabetes, and performed better than clinical features alone (AUC-ROC 0.97 vs. 0.95; P = 0.03). Histological classification of type 1 diabetes was concordant with serum C-peptide [median < 17 pmol/l (limit of detection) vs. 1037 pmol/l in non-type 1 diabetes; P < 0.0001]. CONCLUSIONS: Our study provides robust histological evidence that a clinical diagnostic model, combining clinical features and biomarkers, could improve diabetes classification. Our study also provides reassurance that a C-peptide-based definition of type 1 diabetes is an appropriate surrogate outcome that can be used in large clinical studies where histological definition is impossible. Parts of this study were presented in abstract form at the Network for Pancreatic Organ Donors Conference, Florida, USA, 19-22 February 2019 and Diabetes UK Professional Conference, Liverpool, UK, 6-8 March 2019.


Subject(s)
Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/pathology , Islets of Langerhans/pathology , Adult , Age of Onset , Autoantibodies/immunology , Body Mass Index , C-Peptide/blood , Diabetes Mellitus/classification , Diabetes Mellitus/genetics , Diabetes Mellitus/immunology , Diabetes Mellitus/pathology , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 2/diagnosis , Diagnosis, Differential , Female , Genetic Predisposition to Disease , Humans , Insulin/metabolism , Islets of Langerhans/metabolism , Male , Middle Aged , Pancreas/metabolism , Pancreas/pathology , Reproducibility of Results , Young Adult , Zinc Transporter 8/immunology
2.
Metabolomics ; 15(2): 15, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30830416

ABSTRACT

INTRODUCTION: We recently identified variances in serum metabolomic profiles between fasted diabetic and healthy dogs, some having similarities to those identified in human type 1 diabetes. OBJECTIVES: Compare untargeted metabolomic profiles in the non-fasted state. METHODS: Serum from non-fasted diabetic (n = 6) and healthy control (n = 6) dogs were analyzed by liquid chromatography-high resolution mass spectrometry. RESULTS: Clear clustering of metabolites between groups were observed, with multiple perturbations identified that were similar to those previously observed in fasted diabetic dogs. CONCLUSION: These findings further support the development of targeted assays capable of detecting metabolites that may be useful as biomarkers of canine diabetes.


Subject(s)
Diabetes Mellitus/metabolism , Diabetes Mellitus/veterinary , Dogs/metabolism , Animals , Biomarkers/blood , Chromatography, High Pressure Liquid/methods , Chromatography, Liquid , Cluster Analysis , Dogs/blood , Metabolome , Metabolomics/methods , Tandem Mass Spectrometry
4.
Clin Exp Immunol ; 185(1): 33-41, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27029857

ABSTRACT

The Network for Pancreatic Organ donors with Diabetes (nPOD) programme was developed in response to an unmet research need for human pancreatic tissue obtained from individuals with type 1 diabetes mellitus and people at increased risk [i.e. autoantibody (AAb)-positive] for the disease. This necessitated the establishment of a type 1 diabetes-specific AAb screening platform for organ procurement organizations (OPOs). Assay protocols for commercially available enzyme-linked immunosorbent assays (elisas) determining AAb against glutamic acid decarboxylase (GADA), insulinoma-associated protein-2 (IA-2A) and zinc transporter-8 (ZnT8A) were modified to identify AAb-positive donors within strict time requirements associated with organ donation programmes. These rapid elisas were evaluated by the international islet AAb standardization programme (IASP) and used by OPO laboratories as an adjunct to routine serological tests evaluating donors for organ transplantation. The rapid elisas performed well in three IASPs (2011, 2013, 2015) with 98-100% specificity for all three assays, including sensitivities of 64-82% (GADA), 60-64% (IA-2A) and 62-68% (ZnT8A). Since 2009, nPOD has screened 4442 organ donors by rapid elisa; 250 (5·6%) were identified as positive for one AAb and 14 (0.3%) for multiple AAb with 20 of these cases received by nPOD for follow-up studies (14 GADA+, two IA-2A(+) , four multiple AAb-positive). Rapid screening for type 1 diabetes-associated AAb in organ donors is feasible, allowing for identification of non-diabetic, high-risk individuals and procurement of valuable tissues for natural history studies of this disease.


Subject(s)
Autoantibodies/blood , Donor Selection/standards , Enzyme-Linked Immunosorbent Assay/standards , Tissue Donors/supply & distribution , Adolescent , Adult , Area Under Curve , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/genetics , Cation Transport Proteins/immunology , Child , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/surgery , Female , Glutamate Decarboxylase/antagonists & inhibitors , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/immunology , Humans , Male , Middle Aged , Receptor-Like Protein Tyrosine Phosphatases, Class 8/antagonists & inhibitors , Receptor-Like Protein Tyrosine Phosphatases, Class 8/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 8/immunology , Risk , Sensitivity and Specificity , Zinc Transporter 8
5.
Clin Exp Immunol ; 183(3): 350-7, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26462724

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) has been used to restore immune competence following chemoablative cancer therapy and to promote immunological tolerance in certain settings of autoimmunity. Therefore, we tested the potential of G-CSF to impact type 1 diabetes (T1D) progression in patients with recent-onset disease [n = 14; n = 7 (placebo)] and assessed safety, efficacy and mechanistic effects on the immune system. We hypothesized that pegylated G-CSF (6 mg administered subcutaneously every 2 weeks for 12 weeks) would promote regulatory T cell (Treg) mobilization to a degree capable of restoring immunological tolerance, thus preventing further decline in C-peptide production. Although treatment was well tolerated, G-CSF monotherapy did not affect C-peptide production, glycated haemoglobin (HbA1c) or insulin dose. Mechanistically, G-CSF treatment increased circulating neutrophils during the 12-week course of therapy (P < 0·01) but did not alter Treg frequencies. No effects were observed for CD4(+) : CD8(+) T cell ratio or the ratio of naive : memory (CD45RA(+)/CD45RO(+)) CD4(+) T cells. As expected, manageable bone pain was common in subjects receiving G-CSF, but notably, no severe adverse events such as splenomegaly occurred. This study supports the continued exploration of G-CSF and other mobilizing agents in subjects with T1D, but only when combined with immunodepleting agents where synergistic mechanisms of action have previously demonstrated efficacy towards the preservation of C-peptide.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , Granulocyte Colony-Stimulating Factor/administration & dosage , Immune Tolerance , Insulin-Secreting Cells/physiology , Polyethylene Glycols/administration & dosage , Adolescent , Adult , C-Peptide/blood , CD4-CD8 Ratio , Child , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/physiopathology , Disease Progression , Drug Administration Schedule , Female , Glycated Hemoglobin/analysis , Granulocyte Colony-Stimulating Factor/adverse effects , Granulocyte Colony-Stimulating Factor/blood , Granulocyte Colony-Stimulating Factor/therapeutic use , Humans , Insulin/therapeutic use , Insulin-Secreting Cells/drug effects , Leukocyte Count , Lymphocyte Depletion , Male , Middle Aged , Neutrophils/drug effects , Neutrophils/physiology , Polyethylene Glycols/adverse effects , Polyethylene Glycols/therapeutic use , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/therapeutic use , Splenomegaly , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Young Adult
6.
Immunogenetics ; 65(1): 83-90, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23081744

ABSTRACT

Tyrosine kinase inhibitors (TKi) hold promise as a treatment for a variety of disorders ranging from those in oncology to diseases thought as immune mediated. Tyrphostin AG490 is a potent Jak-Stat TKi shown effective in the prevention of allograft transplant rejection, experimental autoimmune disease, as well as the treatment of cancer. However, given its ability to modulate this important but pleiotropic intracellular pathway, we thought that it is important to examine its effects on glucose metabolism and expression of major transcription factors and adipokines associated with insulin insensitivity and diabetes. We investigated the metabolic effects of AG490 on glucose levels in vivo using an animal model of diabetes, nonobese diabetic (NOD) mice, and transcription factor expression through assessment of human adipocytes. AG490 treatment of young nondiabetic NOD mice significantly reduced blood glucose levels (p = 0.002). In vitro, treatment of adipocytes with rosiglitazone, an insulin sensitizer that binds to peroxisome proliferator-activated receptor (PPAR) receptors and increases the adipocyte response to insulin, significantly increased the expression of the antidiabetic adipokine adiponectin. Importantly, the combination of rosiglitazone plus Tyrphostin AG490 further increased this effect and was specifically associated with significant upregulation of C-enhanced binding protein (C/EBP) (p < 0.0001). In terms of the mechanism underlying this action, regulatory regions of the PPARγ, ADIPOQ, and C/EBP contain the Stat5 DNA-binding sequences and were demonstrated, by gel shift experiments in vitro. These data suggest that blocking Jak-Stat signaling with AG490 reduces blood glucose levels and modulates the expression of transcription factors previously associated with diabetes, thereby supporting its potential as a therapy for this disease.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Diabetes Mellitus, Type 1/metabolism , Enzyme Inhibitors/pharmacology , Thiazolidinediones/pharmacology , Tyrphostins/pharmacology , Adiponectin/metabolism , Animals , Biomarkers/analysis , Blood Glucose/metabolism , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Type 1/drug therapy , Enzyme Inhibitors/administration & dosage , Glucose/analysis , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , Janus Kinases/antagonists & inhibitors , Mice , Mice, Inbred NOD , PPAR gamma/antagonists & inhibitors , PPAR gamma/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Rosiglitazone , Thiazolidinediones/administration & dosage , Tyrphostins/administration & dosage
7.
Diabetologia ; 54(9): 2232-6, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21773685

ABSTRACT

For more than 40 years, the contributions of nurture (i.e. the environment) and nature (i.e. genetics) have been touted for their aetiological importance in type 1 diabetes. Disappointingly, knowledge gains in these areas, while individually successful, have to a large extent occurred in isolation from each other. One reason underlying this divide is the lack of a testable model that simultaneously considers the contributions of genetic and environmental determinants in the formation of this and potentially other disorders that are subject to these variables. To address this void, we have designed a model based on the hypothesis that the aetiological influences of genetics and environment, when evaluated as intersecting and reciprocal trend lines based on odds ratios, result in a method of concurrently evaluating both facets and defining the attributable risk of clinical onset of type 1 diabetes. The model, which we have elected to term the 'threshold hypothesis', also provides a novel means of conceptualising the complex interactions of nurture with nature in type 1 diabetes across various geographical populations.


Subject(s)
Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/genetics , Environment , Models, Biological , Age Factors , Diabetes Mellitus, Type 1/epidemiology , Genetic Predisposition to Disease/etiology , Genetic Predisposition to Disease/genetics , Humans , Incidence , Risk Factors
8.
Scand J Immunol ; 72(4): 293-301, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20883314

ABSTRACT

Our previous studies showed that high levels of soluble CD25 (sCD25) in the serum of patients with hepatocellular carcinoma (HCC) correlated with blunted effector T-cells (Teff) responses, tumour burden and poor survival. Understanding the interactions between Teff, CD4+CD25+ regulatory T cells (Treg) and soluble factors can identify novel therapeutic targets. In this study, we characterize the mechanisms by which HCC serum and sCD25 mediate suppression of Teff and evaluate the effect of sCD25 on the suppression assays with normal healthy control cells (NHC) at a 1:1 Treg to Teff cell ratio to determine whether sCD25 has any impact on Treg suppression. HCC serum and sCD25 suppressed Teff proliferation and downregulated CD25 expression on HCC Teff in a dose-dependent fashion with sCD25 doses above 3000 pg/ml. Treg from HCC and cirrhosis patients suppressed proliferation of target CD4+CD25- Teff in serum-free medium (SFM). HCC Treg showed a higher degree of suppression than cirrhosis-derived Treg. In contrast, Treg from NHC did not suppress target Teff in SFM. However, isolated Treg from all three study subjects (HCC, cirrhosis and NHC) suppressed CD4+CD25- Teff in serum conditions or in the presence of sCD25 in the range 6000-12,000 pg/ml. In conclusion, downregulation of CD25 cell surface expression on Teff is part of the overall suppressive mechanism of sCD25 and HCC serum on Teff responses. The observed sCD25 and HCC serum-mediated suppression is further influenced via novel immune-inhibitory interaction between CD4+CD25+ Treg and sCD25.


Subject(s)
Carcinoma, Hepatocellular/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Liver Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes/immunology , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Cells, Cultured , Culture Media, Serum-Free/pharmacology , Flow Cytometry , Humans , Interleukin-2 Receptor alpha Subunit/blood , Liver Neoplasms/blood , Liver Neoplasms/pathology , Solubility , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism
9.
Diabetologia ; 53(10): 2198-204, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20593162

ABSTRACT

AIMS/HYPOTHESIS: Human alpha1-antitrypsin (hAAT) gene therapy prevents type 1 diabetes in a NOD mouse model of diabetes. However, repeated i.p. injections of hAAT into NOD mice leads to fatal anaphylaxis. The aim of the study was to determine if an alternative route of administration avoids anaphylaxis and allows evaluation of hAAT's potential for diabetes prevention and reversal. We also sought to determine if the addition of granulocyte colony-stimulating factor (G-CSF), augments hAAT's capacity to prevent or reverse disease in the NOD mice. METHODS: To evaluate hAAT pharmacokinetics, serum hAAT levels were monitored in NOD mice receiving a single dose (2 mg) of hAAT by i.p., s.c. or i.d. injection. For studies of type 1 diabetes prevention and reversal, mice received i.d. hAAT (2 mg/mouse/3 days) for 8 or 10 weeks or hAAT and G-CSF (i.p., 6 microg/day) for 6 weeks. Blood glucose determinations, glucose tolerance testing and insulin tolerance tests were performed. RESULTS: Both i.p. and s.c. injections resulted in fatal anaphylaxis. The i.d. injection avoided anaphylaxis and i.d. injection of hAAT into 11-week-old NOD mice prevented disease (p = 0.005, AAT vs PBS at 40 weeks of age). Treatment of diabetic NOD mice with hAAT or hAAT plus G-CSF provided long-term (at least 100 days) reversal of diabetes in 50% of treated animals. G-CSF did not enhance the reversal rates of hAAT. Glucose tolerance and insulin levels were normalised in mice with hAAT prevention and reversal. CONCLUSIONS/INTERPRETATION: Intradermal hAAT prevents and reverses disease in a NOD mouse model of type 1 diabetes without inducing anaphylaxis.


Subject(s)
Anaphylaxis/prevention & control , Diabetes Mellitus, Type 1/drug therapy , Hyperglycemia/drug therapy , alpha 1-Antitrypsin/therapeutic use , Animals , Area Under Curve , Diabetes Mellitus, Type 1/prevention & control , Female , Glucose Intolerance/drug therapy , Glucose Tolerance Test , Hyperglycemia/prevention & control , Immunohistochemistry , Insulin/blood , Mice , Mice, Inbred NOD , alpha 1-Antitrypsin/pharmacokinetics
10.
Diabetologia ; 53(4): 690-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20062967

ABSTRACT

AIMS/HYPOTHESIS: Childhood diabetes is thought to usually result from autoimmune beta cell destruction (type 1A) with eventual total loss of beta cells. Analysis of C-peptide in children characterised at diabetes onset for autoantibodies shows heterogeneous preservation of insulin secretion in long-standing diabetes. The aim of this study was to characterise the pancreases of childhood-onset diabetes in order to define the pathological basis of this heterogeneity. METHODS: We evaluated 20 cadaveric organ donor pancreases of childhood-onset long-term patients for disease heterogeneity and obtained corresponding C-peptide measurements. RESULTS: Pancreases from the majority of cadaveric donors contained only insulin-deficient islets (14 of 20). The remaining six patients (30%) had numerous insulin-positive cells within at least some islets, with two different histological patterns. Pattern A (which we would associate with type 1A diabetes) had lobular retention of areas with 'abnormal' beta cells producing the apoptosis inhibitor survivin and HLA class I. In pattern B, 100% of all islets contained normal-appearing but quantitatively reduced beta cells without survivin or HLA class I. CONCLUSIONS/INTERPRETATION: Our data demonstrate that C-peptide secretion in long-standing diabetic patients can be explained by two different patterns of beta cell survival,possibly reflecting different subsets of type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/pathology , Insulin-Secreting Cells/pathology , Pancreas/pathology , Sex Characteristics , Adolescent , Adult , Age of Onset , Autoantibodies/blood , C-Peptide/blood , Child , Child, Preschool , Diabetes Mellitus, Type 1/immunology , Female , HLA-DR Antigens , Histocompatibility Testing , Humans , Hyperinsulinism/pathology , Male , Middle Aged , Tissue Donors
11.
Diabetologia ; 52(2): 262-70, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19002428

ABSTRACT

AIMS/HYPOTHESIS: Recent histological analysis of pancreases obtained from patients with long-standing type 1 diabetes identified chronic islet inflammation and limited evidence suggestive of beta cell replication. Studies in rodent models also suggest that beta cell replication can be induced by certain inflammatory cytokines and by gastrin. We therefore tested the hypothesis that beta cell replication is observed in non-autoimmune human pancreatic disorders in which localised inflammation or elevated gastrin levels are present. METHODS: Resected operative pancreatic specimens were obtained from patients diagnosed with primary adenocarcinoma (with or without chronic severe pancreatitis) or gastrinoma. Additional pancreatic tissue was obtained from autopsy control patients. Immunohistochemistry was used to assess fractional insulin area, beta cell number and replication rate and differentiation factors relevant to beta cell development. RESULTS: Fractional insulin area was similar among groups. Patients with pancreatic adenocarcinoma and localised chronic severe pancreatitis displayed significant increases in the number of single beta cells, as well as increased beta cell replication rate and levels of neurogenic differentiation 1 in islets. Patients with gastrinoma demonstrated significant increases in the number of single beta cells, but the beta cell replication rate and islet differentiation factor levels were similar to those in the control group. CONCLUSIONS/INTERPRETATION: These findings indicate that chronic severe pancreatic inflammation can be associated with significant effects on beta cell number or replication rate, depending on the distribution of the cells. This information may prove useful for attempts seeking to design therapies aimed at inducing beta cell replication as a means of reversing diabetes.


Subject(s)
Adenocarcinoma/pathology , Insulin-Secreting Cells/pathology , Insulin/analysis , Pancreatic Neoplasms/pathology , Pancreatitis, Chronic/pathology , Adult , Aged , Aged, 80 and over , Autopsy , Cell Nucleus/pathology , Female , Humans , Immunohistochemistry , Ki-67 Antigen/analysis , Male , Middle Aged , Reference Values , Young Adult
12.
Clin Exp Immunol ; 154(1): 15-21, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18759852

ABSTRACT

Previous studies have shown that human alpha-1 antitrypsin (hAAT) gene delivery prevents type 1 diabetes (T1D) in non-obese diabetic (NOD) mice. Furthermore, hAAT protein administration prolongs acceptance of islet allografts. Therefore, we evaluated the use of purified hAAT protein therapy to prevent T1D in NOD mice. Female NOD, non-obese resistant (NOR), Balb/c and C57BL/6 mice were injected intraperitoneally with vehicle alone or vehicle containing hAAT, human albumin or mouse albumin (or mg/injection/mouse; 2x/week). Preparations of clinical-grade hAAT included API(R), Aralast, Prolastin and Zemaira. Surprisingly, hAAT administration was associated with a high rate of fatal anaphylaxis. In studies seeking T1D prevention at 4 weeks of age, 100% mice died after six injections of hAAT. When administrated at 8-10 weeks of age, most (80-100%) NOD mice died following the fourth injection of hAAT, while 0% of Balb/c and C57BL/6 mice and 10% of NOR mice died. Interestingly, repeated injections of human albumin, but not mouse albumin, also induced sudden death in NOD mice. Antibodies to hAAT were induced 2-3 weeks after hAAT administration and death was prevented by treatment with anti-platelet-activating factor along with anti-histamine. In studies of disease reversal in NOD mice, using the four pharmaceutical grade formulations of hAAT, anaphylactic deaths were observed with all hAAT preparations. The propensity for fatal anaphylaxis following antigenic administration appears to be NOD- but not hAAT-specific. The susceptibility of NOD mice to hypersensitivity provides a significant limitation for testing of hAAT. Development of strategies to avoid this unwanted response is required to use this promising therapeutic agent for T1D.


Subject(s)
Anaphylaxis/immunology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , alpha 1-Antitrypsin/adverse effects , Albumins/adverse effects , Animals , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Drug Administration Schedule , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Species Specificity , alpha 1-Antitrypsin/therapeutic use
13.
Transplant Proc ; 40(2): 455-6, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18374099

ABSTRACT

Immunomodulatory properties have been recognized for human alpha-1 antitrypsin (hAAT). However, production of anti-hAAT antibodies in mice may inactivate the protein. In this study, we evaluated the effects of chronic hAAT administration on allogeneic islet graft survival. Chemically diabetic mice lacking an efficient humoral response due to the targeted disruption of the Ig mu-chain (muMT mice) or wild-type (WT) C57BL/6 mice received DBA/2 mouse islets under the kidney capsule. hAAT (Prolastin or Aralast) was given intraperitoneally on day 0 and every 3 days thereafter. Control animals received no treatment. hAAT administration in WT mice resulted in prolongation of islet allograft survival in a dose-dependent fashion in both hAAT-treated groups. Lack of Ig response (muMT mice) per se conferred a beneficial effect on graft survival that worsened in the Prolastin-treated groups but improved in the Aralast-treated group. Our data indicate that systemic administration of hAAT results in prolongation of islet allograft survival. Absence of mature B cells and Ig mu-chain resulted in improved graft survival, pointing to a role for B cells in the rejection process in this model. Treatment with Prolastin worsened graft survival in muMT mice, whereas Aralast did improve it, suggesting a different efficacy and possible actions of the two drug formulations.


Subject(s)
Antibody Formation/physiology , Diabetes Mellitus, Experimental/surgery , Graft Survival/drug effects , Islets of Langerhans Transplantation/physiology , alpha 1-Antitrypsin/therapeutic use , Animals , Antibody Formation/drug effects , Graft Survival/immunology , Islets of Langerhans Transplantation/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Subrenal Capsule Assay , Transplantation, Homologous
14.
Transplant Proc ; 40(2): 457-8, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18374100

ABSTRACT

Alpha-1 antitrypsin (AAT) is a serine protease inhibitor able to prevent diabetes onset in nonobese diabetic (NOD) mice and to prolong islet allograft survival in a nonautoimmune murine model. In this study, we explored the effect of chronic administration of human AAT (hAAT) on allogeneic (C57BL/6) islet graft survival in spontaneously diabetic female NOD mice. Mice received intraperitoneal treatment with saline, Prolastin (1 or 2 mg/mouse) or Aralast (2 mg/mouse) on days -1, 0, 3, 6, and 9. Saline-treated mice rejected the grafts 10.0 +/- 2.5 days after transplantation (n = 9). Prolastin 1 mg (n = 9) and 2 mg (n = 3) resulted in rejection on 8.7 +/- 1.4 (not significant) and 13.0 +/- 4.3 days (P < .03), respectively. Aralast-treated mice showed prolongation of graft survival (13 +/- 5.9 days; n = 5; P < .03). Notably, repeated administrations of either hAAT formulation led to sudden death of a proportion of treated animals. Collectively, our preliminary data indicate that prolongation of islet allograft survival in the stringent autoimmune diabetic NOD mouse model can be achieved with hAAT monotherapy. The death of a proportion of treated animals may be consequent to immunization to hAAT and lethal hypersensitivity. Interestingly, this phenomenon was not observed in a non-autoimmune mouse strain (C57BL/6) despite extended hAAT treatment (>100 days).


Subject(s)
Graft Survival/drug effects , Islets of Langerhans Transplantation/physiology , Serine Proteinase Inhibitors/therapeutic use , alpha 1-Antitrypsin/therapeutic use , Animals , Female , Graft Rejection/prevention & control , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Transplantation, Homologous
15.
Am J Transplant ; 7(5): 1112-20, 2007 May.
Article in English | MEDLINE | ID: mdl-17456199

ABSTRACT

Interleukin-10 (IL-10) is a pleiotropic cytokine that plays a pivotal role in the regulation of immune responses. Hence, we evaluated the effects of a recombinant adeno-associated viral vector 1 (rAAV1) encoding rat IL-10 (rAAV1-IL-10) in a rat model of kidney allograft rejection. Dark Agouti rat kidneys were transplanted into Wistar-Furth (WF) rats 8 weeks following a single intramuscular administration of either rAAV1-IL-10 or rAAV1-green fluorescence protein (GFP). Isografts (WF-WF) served as an additional experimental control. Both allograft and isograft recipients received daily cyclosporine (10 mg/kg) for 14 days after transplantation. Serum IL-10 levels increased at 8, 12 and 16 weeks following vector administration in rAAV1-IL-10-treated animals, but not in rAAV1-GFP and isograft groups. rAAV1-IL-10 treatment resulted in lower BUN and creatinine levels (p<0.001), as well as increased allograft survival rates from 22% to 90%. Allograft histological abnormalities were significantly attenuated in the rAAV1-IL-10-treated rats compared with those of rAAV1-GFP controls. Serum levels of proinflammatory cytokines such as growth-related oncogene were also significantly higher in the rAAV1-GFP group than in the rAAV1-IL-10 group. These data suggest delivery of IL-10 using a rAAV1 vector improves renal function and prolongs graft survival in a rat model of kidney transplant rejection.


Subject(s)
Dependovirus/genetics , Genetic Vectors , Graft Survival/drug effects , Interleukin-10/genetics , Interleukin-10/pharmacology , Kidney Transplantation/physiology , Animals , Blood Urea Nitrogen , Creatinine/metabolism , Cytokines/blood , Female , Graft Rejection/physiopathology , Graft Rejection/prevention & control , Graft Survival/physiology , Green Fluorescent Proteins , Injections, Intramuscular , Interleukin-10/blood , Kidney/drug effects , Kidney/pathology , Kidney/physiology , Kidney Transplantation/pathology , Models, Animal , Rats , Rats, Inbred Strains , Rats, Inbred WF , Transplantation, Homologous
17.
Neurology ; 64(4): 743-5, 2005 Feb 22.
Article in English | MEDLINE | ID: mdl-15728308

ABSTRACT

The pathogenic mechanisms underlying Batten disease are unclear. Patients uniformly possess autoantibodies against glutamic acid decarboxylase (GAD) that are predominantly reactive with a region of GAD (amino acids 1 to 20) distinct from subjects with autoimmune type 1 diabetes or stiff-person syndrome. Batten patients did not possess autoantibodies against other type 1 diabetes-associated autoantigens and human leukocyte antigen genotypes revealed no specific associations with this disease.


Subject(s)
Autoantibodies/immunology , Autoantigens/immunology , Autoimmune Diseases/immunology , Glutamate Decarboxylase/immunology , Neuronal Ceroid-Lipofuscinoses/immunology , Adolescent , Adult , Antibody Specificity , Autoantibodies/analysis , Autoantibodies/blood , Autoimmune Diseases/genetics , Child , Child, Preschool , Diabetes Mellitus, Type 1/immunology , Epitopes/immunology , Female , HLA Antigens/genetics , Humans , Infant , Insulin/immunology , Islets of Langerhans/immunology , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Proteins/immunology , Middle Aged , Molecular Chaperones/genetics , Neuronal Ceroid-Lipofuscinoses/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/immunology , Receptor-Like Protein Tyrosine Phosphatases, Class 8 , Stiff-Person Syndrome/immunology
18.
Diabetologia ; 48(1): 68-74, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15602651

ABSTRACT

AIMS/HYPOTHESIS: Immunological and genetic markers can be used to assess risk of developing type 1 diabetes prior to the onset of clinical symptoms. Autoantibody-positive relatives of patients with type 1 diabetes are at increased risk for disease, while the presence of HLA DQA1*0102/DQB1*0602 is thought to confer protection. Using the unique population identified by the Diabetes Prevention Trial--Type Diabetes (DPT-1), our aim was to determine if these individuals were protected from type 1 diabetes. METHODS: We described metabolic and immunological characteristics of islet cell cytoplasmic autoantibodies-positive relatives with DQB1*0602 identified as part of DPT-1. RESULTS: We found that 32% of DQB1*0602-positive relatives identified through the DPT-1 had abnormalities of glucose tolerance despite the fact that only 19% had multiple type 1 diabetes-associated autoantibodies and only 13% had abnormal insulin secretion, markers typically associated with the disease. In addition, these markers were not associated with abnormal glucose tolerance. In contrast, the DQB1*0602-positive relatives had elevated fasting insulin (117+/-10 pmol/l) and homeostasis model assessment of insulin resistance (HOMA-R) (4.90+/-0.5) values, which are more commonly associated with type 2 diabetes. The later marker of insulin resistance was associated with glucose tolerance status. CONCLUSIONS/INTERPRETATION: Our data indicate that DQA1*0102/DQB1*0602 relatives identified through DPT-1 have a high frequency of abnormal glucose tolerance and a disease phenotype with characteristics of type 1 and type 2 diabetes. Thus, multiple pathways to abnormal glucose tolerance are present within families of these type 1 patients.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Glucose Intolerance/genetics , Glucose Tolerance Test , HLA-DQ Antigens/genetics , Autoantibodies/blood , Blood Glucose/metabolism , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Glucose Intolerance/epidemiology , Glucose Intolerance/immunology , HLA-DQ Antigens/immunology , HLA-DQ alpha-Chains , HLA-DQ beta-Chains , Homeostasis , Humans , Insulin Resistance/genetics , Insulin Resistance/physiology
19.
Gene Ther ; 11(3): 233-40, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14737082

ABSTRACT

Adeno-associated virus (AAV) is widely considered a promising vector for therapeutic gene delivery. This promise is based on previous studies assessing AAVs safety and toxicity, ability to infect nondividing cells, elicit a limited immune response and provide long-term gene expression. However, we now find that earlier studies underappreciated the degree of AAV immunogenicity as well as the extent to which genetic background, through regulation of immune responsiveness, influences the duration of gene expression and thereby the effectiveness of AAV-mediated gene therapy. We evaluated antibody responses in 12 mouse strains to AAV serotype 2 (AAV2) and AAV2-expressed transgene products including green fluorescent protein (GFP), human alpha1-antitrypsin and murine interleukin-10. As expected, all immunocompetent mice administered AAV2 developed serologic evidence of immune responsiveness to the virus. However, a previously unidentified serologic prozone effect was observed suggesting that the concentrations of anti-AAV2 antibodies may have historically been subject to marked underestimation. Furthermore, strains with genetic predisposition to autoimmunity (eg, NOD, NZW, MRL-lpr) specifically imparted a functionally deleterious immune response to AAV-delivered transgene products. These findings suggest that more thorough studies of anti-AAV immunity should be performed, and that genetic predisposition to autoimmunity should be considered when assessing AAV efficacy and safety in humans.


Subject(s)
Antibodies, Viral/biosynthesis , Autoimmunity/genetics , Dependovirus/immunology , Genetic Vectors/immunology , Transgenes/immunology , Animals , Female , Gene Transfer Techniques , Genetic Predisposition to Disease , Genetic Therapy , Green Fluorescent Proteins , Immunity, Cellular , Luminescent Proteins/immunology , Mice , Mice, Inbred Strains , Spleen/immunology
20.
Gene Ther ; 11(2): 181-6, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14712302

ABSTRACT

Type I diabetes results from an autoimmune destruction of the insulin-producing pancreatic beta cells. Although the exact immunologic processes underlying this disease are unclear, increasing evidence suggests that immunosuppressive, immunoregulatory and anti-inflammatory agents can interrupt the progression of the disease. Alpha 1 antitrypsin (AAT) is a multifunctional serine proteinase inhibitor (serpin) that also displays a wide range of anti-inflammatory properties. To test the ability of AAT to modulate the development of type I diabetes, we performed a series of investigations involving recombinant adeno-associated virus vector (rAAV)-mediated gene delivery of human alpha-1 antitrypsin (hAAT) to nonobese diabetic (NOD) mice. Recombinant AAV-expressing hAAT (rAAV2-CB-AT) was administered intramuscularly to 4-week-old female NOD mice (1 x 10(10) i.u./mouse). A single injection of this vector reduced the intensity of insulitis, the levels of insulin autoantibodies, and the frequency of overt type I diabetes (30% (3/10) at 32 weeks of age versus 70% (7/10) in controls). Transgene expression at the injection sites was confirmed by immunostaining. Interestingly, antibodies against hAAT were present in a majority of the vector-injected mice and circulating hAAT was undetectable when assessed 10 weeks postinjection. This study suggests a potential therapeutic role for AAT in preventing type I diabetes as well as the ability of AAV gene therapy-based approaches to ameliorate disease effectively.


Subject(s)
Dependovirus/genetics , Diabetes Mellitus, Type 1/prevention & control , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Transduction, Genetic/methods , alpha 1-Antitrypsin/genetics , Animals , Antibodies/analysis , Autoantibodies/blood , Diabetes Mellitus, Type 1/immunology , Female , Humans , Injections, Intramuscular , Insulin/immunology , Mice , Mice, Inbred NOD , alpha 1-Antitrypsin/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...