Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Front Immunol ; 12: 797172, 2021.
Article in English | MEDLINE | ID: mdl-35095874

ABSTRACT

Antigen-specific immunotherapy is an appealing strategy to preserve beta-cell function in type 1 diabetes, although the approach has yet to meet its therapeutic endpoint. Direct administration of autoantigen into lymph nodes has emerged as an alternative administration route that can improve the efficacy of the treatment. In the first open-label clinical trial in humans, injection of aluminum-formulated glutamic acid decarboxylase (GAD-alum) into an inguinal lymph node led to the promising preservation of C-peptide in patients with recent-onset type 1 diabetes. The treatment induced a distinct immunomodulatory effect, but the response at the cell level has not been fully characterized. Here we used mass cytometry to profile the immune landscape in peripheral blood mononuclear cells from 12 participants of the study before and after 15 months of treatment. The immunomodulatory effect of the therapy included reduction of naïve and unswitched memory B cells, increase in follicular helper T cells and expansion of PD-1+ CD69+ cells in both CD8+ and double negative T cells. In vitro stimulation with GAD65 only affected effector CD8+ T cells in samples collected before the treatment. However, the recall response to antigen after 15 months included induction of CXCR3+ and CD11c+Tbet+ B cells, PD-1+ follicular helper T cells and exhausted-like CD8+ T cells. This study provides a deeper insight into the immunological changes associated with GAD-alum administration directly into the lymph nodes.


Subject(s)
B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 1/etiology , Glutamate Decarboxylase/administration & dosage , Immunomodulation/drug effects , Programmed Cell Death 1 Receptor/metabolism , T Follicular Helper Cells/metabolism , B-Lymphocytes/metabolism , Biomarkers , Diabetes Mellitus, Type 1/metabolism , Disease Susceptibility , Humans , Immunophenotyping , Injections, Intralymphatic , Memory T Cells/immunology , Memory T Cells/metabolism , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , T Follicular Helper Cells/immunology
2.
Front Immunol ; 11: 564921, 2020.
Article in English | MEDLINE | ID: mdl-33162978

ABSTRACT

In spite of intensive treatment Type 1 diabetes leads to serious complications. Preservation of residual beta cell function makes the disease milder, facilitates treatment, prevents complications and increase survival. So far immune interventions have had limited effect, and some serious adverse events and risks. In an open pilot trial we aimed to improve efficacy of GAD-alum treatment using lymph-node administration in combination with oral vitamin D. Here we report the clinical effect and focus on biomarkers for response to treatment. Patients (n = 12) aged 12 to 24 years with recent onset of Type 1 diabetes received 4 µg GAD-alum into lymph-node at day 30, 60, and 90, and oral Vitamin D 2000 U/d, days 1 to 120. Beta cell function was estimated by Mixed Meal Tolerance Tests. GADA, GADA subclasses, GAD65-induced cytokines and proliferation, and T cells markers were analyzed. The treatment was tolerable with no adverse events. Fasting C-peptide and insulin requirement remained stable at 15 months, while HbA1c was lower than baseline. Stimulated C-peptide showed no change at 6 months but declined after 15 months (81% of baseline). Eleven patients remained in partial remission (IDAAC < 9). Patients (n = 9) with better clinical outcome had reduced proportion of IgG1 and increased IgG2, IgG3, and IgG4, increased IL-10 secretion, and reduction of proliferation and CD8+ T cells activation. Patients with poorer clinical response had higher baseline levels of GAD65-induced cytokines and T-cell activation, and an increased ratio of effector/central memory T cells. Intra-lymphatic GAD treatment combined with Vitamin D might preserve beta cell function and improve clinical course in T1D. Patients with less benefit have a different quality of immune response both before and after treatment. Clinical Trial Registration: clinicaltrials.gov, identifier NCT02352974.


Subject(s)
Aluminum Hydroxide/administration & dosage , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , Glutamate Decarboxylase/administration & dosage , Immunity/drug effects , Insulin-Secreting Cells/drug effects , Lymph Nodes/immunology , Vitamin D/administration & dosage , Vitamins/administration & dosage , Administration, Oral , Adolescent , C-Peptide/metabolism , CD8-Positive T-Lymphocytes/immunology , Child , Female , Humans , Immunoglobulin G/metabolism , Injections, Intralymphatic , Interleukin-10/metabolism , Lymphocyte Activation/drug effects , Male , Signal Transduction/drug effects , Treatment Outcome , Young Adult
3.
Contemp Clin Trials ; 82: 93-100, 2019 07.
Article in English | MEDLINE | ID: mdl-31229619

ABSTRACT

BACKGROUND: Evidence suggests that GABA may reduce pancreatic inflammation, protect ß-cells from autoimmune destruction, and potentiate the regeneration of new ß-cells in the setting of type 1 diabetes mellitus (T1DM). The enzyme GAD, also expressed in human pancreatic ß-cells, is an antigenic target of reactive T cells. We hypothesized that treatment of children with recent onset T1DM with GABA or combination GABA with GAD will preserve ß-cell function and ameliorate autoimmune dysregulation. METHODS: This is a one-year, prospective, randomized, double-blind, placebo-controlled trial. Ninety-nine patients aged 4-18 years with newly diagnosed T1DM are randomized into three treatment groups: 1) oral GABA twice daily in addition to two injections of recombinant GAD enzyme, 2) oral GABA plus placebo GAD injections, or 3) placebo GABA and placebo GAD. Patients are evaluated at baseline and months 1, 5, 8 and 12. Mixed meal tolerance testing is performed at all but the 8-month visit. Laboratory studies will assess indices of beta and alpha cell function, glycemic control, immunophenotyping, and diabetes-related autoantibodies. RESULTS: The primary outcome is the effect on pancreatic ß-cell function as measured by meal-stimulated c-peptide secretion compared between the treatment groups before and after one year of treatment. Secondary outcomes include: 1) fasting and meal stimulated glucagon and proinsulin levels, 2) response in insulin usage by participants, 3) indices of immune cell function, and 4) effect on autoantibodies GAD65, ICA512, and ZnT8. CONCLUSIONS: This trial will determine the safety and efficacy of GABA and combination GABA/GAD therapy to delay T1DM progression in children.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Glutamate Decarboxylase/therapeutic use , gamma-Aminobutyric Acid/therapeutic use , Adolescent , Child , Child, Preschool , Disease Progression , Double-Blind Method , Drug Therapy, Combination , Female , Glutamate Decarboxylase/administration & dosage , Humans , Injections, Subcutaneous , Male , Randomized Controlled Trials as Topic , gamma-Aminobutyric Acid/administration & dosage
5.
Clin Immunol ; 176: 114-121, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28131926

ABSTRACT

Administration of Glutamic Acid Decarboxylase (GAD)65 formulated in aluminium hydroxide preserved insulin secretion in a phase II trial in recent onset Type 1 Diabetes. A subsequent European phase III trial was closed at 15months after failing to reach primary endpoint, but the majority of the Swedish patients completed the 21months follow-up. We studied the frequencies and phenotype of T cells, suppressive capacity of Tregs, GAD65-induced proliferation, and frequencies of T cells with a GAD65-specific TCR in Swedes participating in the trial. Stimulation with GAD65 induced activated T cells and also cells with a suppressive phenotype. Activated GAD65-specific effector T cells were detected by tetramer staining while the frequency of GAD65-specific Treg was not affected by the treatment. Additional doses of GAD-alum increased frequencies of CD25+CD127+, but had no effect on CD25hiCD127lo. Our findings indicate that GAD-alum treatment primarily induced activated T cells. GAD65-specific cells were mainly of activated phenotype.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , Glutamate Decarboxylase/administration & dosage , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Adolescent , Adult , Child , Double-Blind Method , Female , Follow-Up Studies , Humans , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-7 Receptor alpha Subunit/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Male , Sweden , Young Adult
6.
Epilepsy Behav ; 71(Pt B): 238-242, 2017 06.
Article in English | MEDLINE | ID: mdl-27993512

ABSTRACT

Herpes virus technology involving manipulation of GAD65 was used to study effects on audiogenic seizures (AGS). Audiogenic seizure behaviors were examined following injections of replication-defective herpes simplex virus (HSV-1) vectors incorporating sense or antisense toward GAD65 along with 10% lac-Z into the central nucleus of inferior colliculus (CNIC) of Long-Evans rats. In seizure-sensitive animals developmentally primed by intense sound exposure, injection of GAD65 in the sense orientation increased wild running latencies and reduced incidence of clonus compared with lac-Z only, unoperated, and vehicle seizure groups. In contrast, infection of CNIC with GAD65 antisense virus resulted in 100% incidence of wild running and clonus behaviors in AGS animals. Unprimed animals not operated continued to show uniform absence of seizure activity. Administration of GAD65 antisense virus into CNIC produced novel wild running and clonus behaviors in some unprimed animals. Staining for ß-galactosidase in all vector animals revealed no differences in pattern or numbers of immunoreactive cells at injection sites. Qualitatively, typical small and medium multipolar/stellate and medium fusiform neurons appeared in the CNIC of vector animals. These results demonstrate that HSV-1 vector constructs implanted into the CNIC can predictably influence incidence and severity of AGS and suggest that viral vectors can be useful in studying GABA mechanisms with potential for therapeutic application in epilepsy. This article is part of a Special Issue entitled "Genetic and Reflex Epilepsies, Audiogenic Seizures and Strains: From Experimental Models to the Clinic".


Subject(s)
Acoustic Stimulation/adverse effects , Epilepsy, Reflex/chemically induced , Glutamate Decarboxylase/toxicity , Herpesvirus 1, Human , Inferior Colliculi/drug effects , Seizures/chemically induced , Animals , Epilepsy, Reflex/pathology , Epilepsy, Reflex/physiopathology , Female , Glutamate Decarboxylase/administration & dosage , Inferior Colliculi/pathology , Inferior Colliculi/physiopathology , Male , Neurons/drug effects , Neurons/pathology , Rats , Rats, Long-Evans , Seizures/physiopathology
7.
PLoS One ; 11(1): e0147260, 2016.
Article in English | MEDLINE | ID: mdl-26783749

ABSTRACT

Induction of mucosal tolerance by oral administration of protein antigens is a potential therapeutic strategy for preventing and treating type 1 diabetes (T1D); however, the requirement for a large dosage of protein limits clinical applications because of the low efficacy. In this study, we generated a fusion protein CTB-Ins-GAD composed of CTB (cholera toxin B subunit), insulin, and three copies of GAD65 peptide 531-545, which were efficiently produced in silkworm pupae, to evaluate its protective effect against T1D. We demonstrate that oral administration of CTB-Ins-GAD suppressed T1D by up to 78%, which is much more effective than GAD65 single-antigen treatment. Strikingly, CTB-Ins-GAD enhance insulin- and GAD65-specific Th2-like immune responses, which repairs the Th1/Th2 imbalance and increases the number of CD4(+)CD25(+)Foxp3(+) T cell and suppresses insulin- and GAD65-reactive spleen T lymphocyte proliferation and migration. Our results strongly suggest that the combined dual antigens promote the induction of oral tolerance, thus providing an effective and economic immunotherapy against T1D in combination with a silkworm bioreactor.


Subject(s)
Autoantigens/administration & dosage , Bombyx/enzymology , Diabetes Mellitus, Type 1/prevention & control , Glutamate Decarboxylase/administration & dosage , Insulin/administration & dosage , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Administration, Oral , Adoptive Transfer , Animals , Autoantigens/immunology , Blotting, Western , Cell Differentiation , Cholera Toxin/administration & dosage , Cholera Toxin/immunology , Diabetes Mellitus, Type 1/epidemiology , Diabetes Mellitus, Type 1/immunology , Female , Glutamate Decarboxylase/immunology , Incidence , Insulin/immunology , Mice, Inbred NOD , Mice, SCID , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/biosynthesis
9.
Diabetes ; 63(8): 2876-87, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24677716

ABSTRACT

Growing insight into the pathogenesis of type 1 diabetes (T1D) and numerous studies in preclinical models highlight the potential of antigen-specific approaches to restore tolerance efficiently and safely. Oral administration of protein antigens is a preferred method for tolerance induction, but degradation during gastrointestinal passage can impede such protein-based therapies, reducing their efficacy and making them cost-ineffective. To overcome these limitations, we generated a tolerogenic bacterial delivery technology based on live Lactococcus lactis (LL) bacteria for controlled secretion of the T1D autoantigen GAD65370-575 and the anti-inflammatory cytokine interleukin-10 in the gut. In combination with short-course low-dose anti-CD3, this treatment stabilized insulitis, preserved functional ß-cell mass, and restored normoglycemia in recent-onset NOD mice, even when hyperglycemia was severe at diagnosis. Combination therapy did not eliminate pathogenic effector T cells, but increased the presence of functional CD4(+)Foxp3(+)CD25(+) regulatory T cells. These preclinical data indicate a great therapeutic potential of orally administered autoantigen-secreting LL for tolerance induction in T1D.


Subject(s)
Autoantigens/pharmacology , Diabetes Mellitus/immunology , Glutamate Decarboxylase/pharmacology , Interleukin-10/metabolism , Peptide Fragments/pharmacology , Administration, Oral , Aging , Animals , Autoantigens/administration & dosage , Autoantigens/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Glutamate Decarboxylase/administration & dosage , Interleukin-10/genetics , Lactococcus lactis , Mice , Mice, Inbred NOD , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , T-Lymphocytes, Regulatory/drug effects
10.
PLoS One ; 8(9): e72921, 2013.
Article in English | MEDLINE | ID: mdl-24058450

ABSTRACT

Stiff person syndrome (SPS) is a highly-disabling neurological disorder of the CNS characterized by progressive muscular rigidity and spasms. In approximately 60-80% of patients there are autoantibodies to glutamic acid decarboxylase (GAD), the enzyme that synthesizes gamma-amino butyric acid (GABA), the predominant inhibitory neurotransmitter of the CNS. Although GAD is intracellular, it is thought that autoimmunity to GAD65 may play a role in the development of SPS. To test this hypothesis, we immunized mice, that expressed enhanced green fluorescent protein (EGFP) under the GAD65 promoter, with either GAD65 (n = 13) or phosphate buffered saline (PBS) (n = 13). Immunization with GAD65 resulted in autoantibodies that immunoprecipitated GAD, bound to CNS tissue in a highly characteristic pattern, and surprisingly bound not only to GAD intracellularly but also to the surface of cerebellar neurons in culture. Moreover, immunization resulted in immunoglobulin diffusion into the brainstem, and a partial loss of GAD-EGFP expressing cells in the brainstem. Although immunization with GAD65 did not produce any behavioral abnormality in the mice, the induction of neuronal-surface antibodies and the trend towards loss of GABAergic neurons in the brainstem, supports a role for humoral autoimmunity in the pathogenesis of SPS and suggests that the mechanisms may involve spread to antigens expressed on the surface of these neurons.


Subject(s)
Autoantibodies/biosynthesis , Cerebellum/immunology , GABAergic Neurons/immunology , Glutamate Decarboxylase/administration & dosage , Mutant Chimeric Proteins/immunology , Stiff-Person Syndrome/immunology , Animals , Autoantibodies/immunology , Autoimmunity , Cells, Cultured , Cerebellum/pathology , Female , GABAergic Neurons/pathology , Genes, Reporter , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/immunology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunization , Male , Mice , Mice, Transgenic , Mutant Chimeric Proteins/genetics , Protein Binding , Protein Transport , Stiff-Person Syndrome/chemically induced , Stiff-Person Syndrome/pathology , gamma-Aminobutyric Acid/metabolism
11.
Diabetes Care ; 36(11): 3418-24, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23863909

ABSTRACT

OBJECTIVE: GAD formulated in aluminum hydroxide (GAD-alum) has previously been shown to induce preservation of residual insulin secretion in recent-onset type 1 diabetes, but recent phase II and III GAD-alum trials failed to reach primary outcomes. The European phase III study was therefore closed after 15 months, and only a minority of patients completed the 30 months of follow-up. RESEARCH DESIGN AND METHODS: This study aimed to characterize cellular and humoral responses in the Swedish patients (n = 148) participating in the phase III trial, receiving four (4D) or two (2D) GAD-alum doses or placebo. Serum GAD65 antibody (GADA) levels, GADA IgG1-4 subclass distribution, cytokine secretion, and proliferative responses in peripheral blood mononuclear cells (PBMCs) were analyzed. RESULTS: The GAD65-induced cytokine profile tended to switch toward a predominant Th2-associated profile over time both in the 2D and 4D group. The groups also displayed increased GADA levels and PBMC proliferation compared with placebo, whereas GADA IgG subclass distribution changed in 4D patients. CONCLUSIONS: Both 2D and 4D patients displayed GAD65-specifc cellular and humoral effects after GAD-alum treatment, but at different time points and magnitudes. No specific immune markers could be associated with treatment efficacy.


Subject(s)
Alum Compounds/administration & dosage , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , Glutamate Decarboxylase/administration & dosage , Immunity, Cellular , Immunity, Humoral , Adult , Autoantibodies/blood , Female , Glutamate Decarboxylase/immunology , Humans , Immunoglobulin G/blood , Insulin/therapeutic use , Leukocytes, Mononuclear/immunology , Male , Middle Aged
12.
Clin Exp Immunol ; 172(3): 394-402, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23600827

ABSTRACT

Glutamic acid decarboxylase (GAD)(65) formulated with aluminium hydroxide (GAD-alum) was effective in preserving insulin secretion in a Phase II clinical trial in children and adolescents with recent-onset type 1 diabetes. In addition, GAD-alum treated patients increased CD4(+) CD25(hi) forkhead box protein 3(+) (FoxP3(+)) cell numbers in response to in-vitro GAD(65) stimulation. We have carried out a 4-year follow-up study of 59 of the original 70 patients to investigate long-term effects on the frequency and function of regulatory T cells after GAD-alum treatment. Peripheral blood mononuclear cells were stimulated in vitro with GAD65 for 7 days and expression of regulatory T cell markers was measured by flow cytometry. Regulatory T cells (CD4(+) CD25(hi) CD127(lo)) and effector T cells (CD4(+) CD25(-) CD127(+)) were further sorted, expanded and used in suppression assays to assess regulatory T cell function after GAD-alum treatment. GAD-alum-treated patients displayed higher frequencies of in-vitro GAD(65) -induced CD4(+) CD25(+) CD127(+) as well as CD4(+) CD25(hi) CD127(lo) and CD4(+) FoxP3(+) cells compared to placebo. Moreover, GAD(65) stimulation induced a population of CD4(hi) cells consisting mainly of CD25(+) CD127(+) , which was specific of GAD-alum-treated patients (16 of 25 versus one of 25 in placebo). Assessment of suppressive function in expanded regulatory T cells revealed no difference between GAD-alum- and placebo-treated individuals. Regulatory T cell frequency did not correlate with C-peptide secretion throughout the study. In conclusion, GAD-alum treatment induced both GAD(65) -reactive CD25(+) CD127(+) and CD25(hi) CD127(lo) cells, but no difference in regulatory T cell function 4 years after GAD-alum treatment.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Glutamate Decarboxylase/administration & dosage , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , Adjuvants, Immunologic/administration & dosage , Adolescent , Alum Compounds/administration & dosage , Autoantigens/administration & dosage , Child , Diabetes Mellitus, Type 1/enzymology , Female , Follow-Up Studies , Glutamate Decarboxylase/immunology , Humans , Immunosuppression Therapy , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-7 Receptor alpha Subunit/metabolism , Lymphocyte Activation , Male , T-Lymphocyte Subsets/enzymology , T-Lymphocyte Subsets/immunology , Time Factors , Treatment Outcome
13.
Clin Exp Immunol ; 171(3): 247-54, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23379430

ABSTRACT

Previous studies have indicated phenotypical differences in glutamic acid decarboxylase 65 autoantibodies (GADA) found in type 1 diabetes (T1D) patients, individuals at risk of developing T1D and stiff-person syndrome (SPS) patients. In a Phase II trial using aluminium-formulated GAD(65) (GAD-alum) as an immunomodulator in T1D, several patients responded with high GADA titres after treatment, raising concerns as to whether GAD-alum could induce GADA with SPS-associated phenotypes. This study aimed to analyse GADA levels, immunoglobulin (Ig)G1-4 subclass frequencies, b78- and b96·11-defined epitope distribution and GAD(65) enzyme activity in sera from four cohorts with very high GADA titres: T1D patients (n = 7), GAD-alum-treated T1D patients (n = 9), T1D high-risk individuals (n = 6) and SPS patients (n = 12). SPS patients showed significantly higher GADA levels and inhibited the in-vitro GAD(65) enzyme activity more strongly compared to the other groups. A higher binding frequency to the b78-defined epitope was found in the SPS group compared to T1D and GAD-alum individuals, whereas no differences were detected for the b96·11-defined epitope. GADA IgG1-4 subclass levels did not differ between the groups, but SPS patients had higher IgG2 and lower IgG4 distribution more frequently. In conclusion, the in-vitro GADA phenotypes from SPS patients differed from the T1D- and high-risk groups, and GAD-alum treatment did not induce SPS-associated phenotypes. However, occasional overlap between the groups exists, and caution is indicated when drawing conclusions to health or disease status.


Subject(s)
Autoantibodies/immunology , Diabetes Mellitus, Type 1/immunology , Glutamate Decarboxylase/immunology , Adolescent , Adult , Aged , Alum Compounds/administration & dosage , Autoantibodies/blood , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/drug therapy , Female , Glutamate Decarboxylase/administration & dosage , Glutamate Decarboxylase/therapeutic use , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Male , Middle Aged , Phenotype , Stiff-Person Syndrome/etiology , Stiff-Person Syndrome/immunology
14.
PLoS One ; 7(1): e30561, 2012.
Article in English | MEDLINE | ID: mdl-22291989

ABSTRACT

BACKGROUND: Loss of GABA-mediated pre-synaptic inhibition after spinal injury plays a key role in the progressive increase in spinal reflexes and the appearance of spasticity. Clinical studies show that the use of baclofen (GABA(B) receptor agonist), while effective in modulating spasticity is associated with major side effects such as general sedation and progressive tolerance development. The goal of the present study was to assess if a combined therapy composed of spinal segment-specific upregulation of GAD65 (glutamate decarboxylase) gene once combined with systemic treatment with tiagabine (GABA uptake inhibitor) will lead to an antispasticity effect and whether such an effect will only be present in GAD65 gene over-expressing spinal segments. METHODS/PRINCIPAL FINDINGS: Adult Sprague-Dawley (SD) rats were exposed to transient spinal ischemia (10 min) to induce muscle spasticity. Animals then received lumbar injection of HIV1-CMV-GAD65 lentivirus (LVs) targeting ventral α-motoneuronal pools. At 2-3 weeks after lentivirus delivery animals were treated systemically with tiagabine (4, 10, 20 or 40 mg/kg or vehicle) and the degree of spasticity response measured. In a separate experiment the expression of GAD65 gene after spinal parenchymal delivery of GAD65-lentivirus in naive minipigs was studied. Spastic SD rats receiving spinal injections of the GAD65 gene and treated with systemic tiagabine showed potent and tiagabine-dose-dependent alleviation of spasticity. Neither treatment alone (i.e., GAD65-LVs injection only or tiagabine treatment only) had any significant antispasticity effect nor had any detectable side effect. Measured antispasticity effect correlated with increase in spinal parenchymal GABA synthesis and was restricted to spinal segments overexpressing GAD65 gene. CONCLUSIONS/SIGNIFICANCE: These data show that treatment with orally bioavailable GABA-mimetic drugs if combined with spinal-segment-specific GAD65 gene overexpression can represent a novel and highly effective anti-spasticity treatment which is associated with minimal side effects and is restricted to GAD65-gene over-expressing spinal segments.


Subject(s)
GABA Agonists/therapeutic use , Genetic Therapy , Glutamate Decarboxylase/genetics , Muscle Spasticity/therapy , Spine/metabolism , Animals , Anticonvulsants/administration & dosage , Anticonvulsants/adverse effects , Anticonvulsants/therapeutic use , Cells, Cultured , Combined Modality Therapy , Embryo, Mammalian , Female , GABA Agonists/administration & dosage , GABA Agonists/adverse effects , Gene Expression Regulation/physiology , Genetic Therapy/methods , Glutamate Decarboxylase/administration & dosage , Glutamate Decarboxylase/adverse effects , Injections, Spinal , Male , Muscle Spasticity/drug therapy , Muscle Spasticity/genetics , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/adverse effects , Neuroprotective Agents/therapeutic use , Nipecotic Acids/administration & dosage , Nipecotic Acids/adverse effects , Nipecotic Acids/therapeutic use , Rats , Rats, Sprague-Dawley , Spine/pathology , Swine , Swine, Miniature , Tiagabine
16.
Hum Vaccin ; 7(1): 45-9, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21263221

ABSTRACT

Type 1 diabetes is a serious chronic disease in which the pancreatic islet beta cells are destroyed by autoimmunity specifically directed to intracellular autoantigens. Still undefined environmental factors are likely to initiate the disease process. One of the autoantigens is glutamic acid decarboxylase (GAD65) and attempts are made to induce immunological tolerance against this autoantigen. Alum-formulated GAD65 (Diamyd (®)) has been given subcutaneously in two injections with one month apart to recent onset type 1 diabetes patients with positive GAD65 autoantibodies. The injections were found to preserve residual ß-cell function without treatment related serious adverse events. Phase III studies in children with recent onset type 1 diabetes are ongoing along with a study (DIAPREV-IT) aimed at testing whether Diamyd (®) may prevent the clinical onset of diabetes in non-diabetic children with GAD65 autoantibodies and at least one more islet autoantibody. Future studies may include investigation of Diamyd (®) in combination with other immunomodulating autoantigens.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Alum Compounds/administration & dosage , Diabetes Mellitus, Type 1/prevention & control , Glutamate Decarboxylase/administration & dosage , Glutamate Decarboxylase/immunology , Immune Tolerance , Autoimmune Diseases/immunology , Autoimmune Diseases/prevention & control , Clinical Trials, Phase III as Topic , Diabetes Mellitus, Type 1/immunology , Humans , Immunization, Secondary/methods , Injections, Subcutaneous , Vaccination/methods
17.
Clin Immunol ; 138(1): 117-26, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21044870

ABSTRACT

Type 1 diabetes results from autoimmune destruction of insulin producing pancreatic ß-cells. We have shown that treatment with alum-formulated glutamic acid decarboxylase 65 (GAD-alum) preserved residual insulin secretion and induced antigen-specific responses in children with recent onset type 1 diabetes. The aim of this study was to further investigate the immunomodulatory effect of GAD-alum, focusing on CD4(+)CD25(high) cells and their association to cytokine secretion. Samples obtained 21 and 30months after the initial injection of GAD-alum or placebo were included in the present study. GAD(65)-stimulation enhanced the percentage of CD4(+)CD25(high)FOXP3(+) cells, but reduced the percentage of CD4(+)CD25(+) cells, in samples from the GAD-alum treated group. Further, the GAD(65)-induced secretion of IL-5, -10, and -13 correlated with the expression of CD4(+)CD25(high)FOXP3(+) cells, but inversely with CD4(+)CD25(+) cells. These new data suggest that GAD-alum treatment induced GAD(65)-specific T cells with regulatory features.


Subject(s)
Alum Compounds , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/therapy , Forkhead Transcription Factors/metabolism , Glutamate Decarboxylase/immunology , Glutamate Decarboxylase/therapeutic use , Interleukin-2 Receptor alpha Subunit/metabolism , T-Lymphocytes, Regulatory/immunology , Adolescent , Alum Compounds/administration & dosage , Autoantibodies/blood , Autoantibodies/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Count , Child , Diabetes Mellitus, Type 1/enzymology , Diabetes Mellitus, Type 1/immunology , Female , Forkhead Transcription Factors/genetics , Gene Expression/genetics , Gene Expression/immunology , Glutamate Decarboxylase/administration & dosage , Humans , Immunosuppression Therapy/methods , Interferon-gamma/metabolism , Interleukins/metabolism , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation/immunology , Male , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Transforming Growth Factor beta/genetics , Treatment Outcome , Tumor Necrosis Factor-alpha/metabolism
18.
J Urol ; 184(3): 1204-10, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20663524

ABSTRACT

PURPOSE: We investigated whether replication defective herpes simplex virus vectors encoding genes of glutamic acid decarboxylase, the gamma-aminobutyric acid synthesis enzyme, could suppress detrusor-sphincter dyssynergia in rats with spinal cord injury. MATERIALS AND METHODS: One week after spinalization herpes simplex virus vectors expressing glutamic acid decarboxylase and green fluorescent protein were injected into the bladder wall. Spinal cord injured rats without herpes simplex virus injection (sham treated) and those injected with LacZ encoding herpes simplex virus vectors served as controls. Three weeks after viral injection we simultaneously recorded urethral and intravesical pressure in awake rats. RESULTS: In the glutamic acid decarboxylase group the urethral pressure increase during bladder contraction was significantly decreased by 77% to 79% compared with that in the sham treated and LacZ groups. Bladder activity and urethral baseline pressure did not differ among the 3 groups. Intrathecal application of the gamma-aminobutyric acid-A receptor antagonist bicuculline almost completely reversed the decrease in the urethral pressure increase during bladder contractions while intrathecal saclofen (Tocris Cookson, Ellisville, Missouri), a gamma-aminobutyric acid-B receptor antagonist, partially reversed it. In the glutamic acid decarboxylase group the mRNA of glutamic acid decarboxylase 67 was significantly increased in L6-S1 dorsal root ganglia, which is where bladder afferents originate, compared with that in the LacZ group. CONCLUSIONS: Herpes simplex virus based glutamic acid decarboxylase gene transfer to bladder afferent pathway may represent a novel approach to detrusor-sphincter dyssynergia in cases of spinal cord injury.


Subject(s)
Gene Transfer Techniques , Glutamate Decarboxylase/administration & dosage , Spinal Cord Injuries/complications , Urinary Bladder, Neurogenic/etiology , Urinary Bladder, Neurogenic/therapy , Animals , Female , Rats , Rats, Sprague-Dawley , Simplexvirus
19.
J Immunol ; 183(7): 4809-16, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19752238

ABSTRACT

Tissue-specific autoimmune diseases such as type 1 diabetes (T1D) are characterized by T cell-driven pathology. Administration of autoantigenic peptides provides a strategy to selectively target the pathogenic T cell response. Indeed, treatment with beta cell peptides effectively prevents T1D in NOD mice. However, the efficacy of peptide immunotherapy generally wanes as beta cell autoimmunity progresses and islet inflammation increases. With the goal of enhancing the efficacy of peptide immunotherapy, soluble (s)IA(g7)-Ig dimers covalently linked to beta cell autoantigen-derived peptides were tested for the capacity to suppress late preclinical T1D. NOD female mice with established beta cell autoimmunity were vaccinated i.v. with a short course of sIA(g7)-Ig dimers tethered to peptides derived from glutamic acid decarboxylase (GAD)65 (sIA(g7)-pGAD65). Treatment with sIA(g7)-pGAD65 dimers and the equivalent of only approximately 7 microg of native peptide effectively blocked the progression of insulitis and the development of diabetes. Furthermore, suppression of T1D was dependent on beta cell-specific IL-10-secreting CD4+ T cells, although the frequency of GAD65-specific FoxP3-expressing CD4+ T cells was also increased in sIA(g7)-pGAD65 dimer vaccinated NOD mice. These results demonstrate that MHC class II-Ig dimer vaccination is a robust approach to suppress ongoing T cell-mediated autoimmunity, and may provide a superior strategy of adjuvant-free peptide-based immunotherapy to induce immunoregulatory T cells.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class II/administration & dosage , Histocompatibility Antigens Class II/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Cell Differentiation/immunology , Diabetes Mellitus, Type 1/pathology , Dimerization , Epitopes, T-Lymphocyte/genetics , Female , Glutamate Decarboxylase/administration & dosage , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/immunology , Histocompatibility Antigens Class II/genetics , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Peptide Fragments/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Regulatory/pathology , T-Lymphocytes, Regulatory/transplantation , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
20.
Diabetologia ; 52(7): 1363-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19404608

ABSTRACT

AIMS/HYPOTHESIS: The aim of this study was to ascertain whether treatment of GAD65 autoantibody (GADA)-positive diabetic patients with alum-formulated recombinant GAD65 (GAD-alum) is safe and does not compromise beta cell function. METHODS: This Phase 2, placebo-controlled, dose-escalation clinical trial, which was randomized through a central office, was performed in 47 GADA-positive type 2 diabetic patients, who received subcutaneous injections of GAD-alum (4 [n = 9], 20 [n = 8], 100 [n = 9] or 500 [n = 8] microg) or placebo (n = 13) at weeks 1 and 4 of the trial. Participants and caregivers were blinded to group assignments. The primary outcome was safety as assessed by neurological tests, medications and beta cell function evaluated over 5 years, representing the end of the trial. RESULTS: No severe study-related adverse events occurred during the 5 year follow-up. None of the dose groups was associated with an increased risk of starting insulin treatment compared with the placebo group. The use of oral hypoglycaemic agents did not differ between the dose groups. After 5 years, fasting C-peptide levels declined in the placebo group (-0.24; 95% CI -0.41 to -0.07 log(10) nmol/l; p = 0.01) and the 500 microg dose group (-0.37; 95% CI -0.57 to -0.17 log(10) nmol/l; p = 0.003), but not in the 4 microg (-0.10; 95% CI -0.28 to 0.07 log(10) nmol/l; p = 0.20), 20 microg (0.04; 95% CI -0.12 to 0.19 log(10) nmol/l; p = 0.58) and 100 microg (0.00; 95% CI -0.20 to -0.20 log(10) nmol/l; p = 0.98) dose groups. CONCLUSIONS/INTERPRETATION: The primary outcome of safety was achieved, since no severe study-related adverse events occurred. TRIAL REGISTRATION: Because the study was initiated before 1 July 2005, the protocol was not registered in a registry.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/immunology , Glutamate Decarboxylase/administration & dosage , Glutamate Decarboxylase/immunology , Vaccines, Synthetic/administration & dosage , Administration, Oral , Alum Compounds/administration & dosage , Autoantibodies/blood , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Dose-Response Relationship, Drug , Follow-Up Studies , Glutamate Decarboxylase/adverse effects , Humans , Hypoglycemic Agents/administration & dosage , Injections, Subcutaneous , Insulin/administration & dosage , Insulin-Secreting Cells/immunology , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...