Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Neurotoxicol Teratol ; 101: 107320, 2024.
Article in English | MEDLINE | ID: mdl-38199312

ABSTRACT

INTRODUCTION: Methylmercury (MeHg) is an environmental contaminant that is of particular concern in Northern Arctic Canadian populations. Specifically, organic mercury compounds such as MeHg are potent toxicants that affect multiple bodily systems including the nervous system. Developmental exposure to MeHg is a major concern, as the developing fetus and neonate are thought to be especially vulnerable to the toxic effects of MeHg. The objective of this study was to examine developmental exposure to low doses of MeHg and effects upon the adult central nervous system (CNS). The doses of MeHg chosen were scaled to be proportional to the concentrations of MeHg that have been reported in human maternal blood samples in Northern Arctic Canadian populations. METHOD: Offspring were exposed to MeHg maternally where pregnant Sprague Dawley rats were fed cookies that contained MeHg or vehicle (vehicle corn oil; MeHg 0.02 mg/kg/body weight or 2.0 mg/kg/body weight) daily, throughout gestation (21 days) and lactation (21 days). Offspring were not exposed to MeHg after the lactation period and were euthanized on postnatal day 450. Brains were extracted, fixed, frozen, and sectioned for immunohistochemical analysis. A battery of markers of brain structure and function were selected including neuronal GABAergic enzymatic marker glutamic acid decarboxylase-67 (GAD67), apoptotic/necrotic marker cleaved caspase-3 (CC3), catecholamine marker tyrosine hydroxylase (TH), immune inflammatory marker microglia (Cd11b), endothelial cell marker rat endothelial cell antigen-1 (RECA-1), doublecortin (DCX), Bergmann glia (glial fibrillary acidic protein (GFAP)), and general nucleic acid and cellular stains Hoechst, and cresyl violet, respectively. Oxidative stress marker lipofuscin (autofluorescence) was also assessed. Both male and female offspring were included in analysis. Two-way analysis of variance (ANOVA) was utilized where sex and treatment were considered as between-subject factors (p* <0.05). ImageJ was used to assess immunohistochemical results. RESULTS: In comparison with controls, adult rat offspring exposed to both doses of MeHg were observed to have (1) increased GAD67 in the cerebellum; (2) decreased lipofuscin in the locus coeruleus; and (3) decreased GAD67 in the anterior CA1 region. Furthermore, in the substantia nigra and periaqueductal gray, adult male offspring consistently had a larger endothelial cell and capillary perimeter in comparison to females. The maternal high dose of MeHg influenced RECA-1 immunoreactivity in both the substantia nigra and periaqueductal gray of adult rat offspring, where the latter neuronal region also showed statistically significant decreases in RECA-1 immunoreactivity at the maternal low dose exposure level. Lastly, males exposed to high doses of MeHg during development exhibited a statistically significant increase in the perimeter of endothelial cells and capillaries (RECA-1) in the cerebellum, in comparison to male controls. CONCLUSION: Findings suggest that in utero and early postnatal exposure to MeHg at environmentally relevant doses leads to long-lasting and selective changes in the CNS. Exposure to MeHg at low doses may affect GABAergic homeostasis and vascular integrity of the CNS. Such changes may contribute to neurological disturbances in learning, cognition, and memory that have been reported in epidemiological studies.


Subject(s)
Methylmercury Compounds , Prenatal Exposure Delayed Effects , Pregnancy , Rats , Animals , Male , Female , Humans , Methylmercury Compounds/toxicity , Rats, Sprague-Dawley , Glutamate Decarboxylase/metabolism , Glutamate Decarboxylase/pharmacology , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/metabolism , Capillaries/metabolism , Endothelial Cells/metabolism , Lipofuscin/metabolism , Lipofuscin/pharmacology , Canada , Cerebellum , Mesencephalon/metabolism , Body Weight
2.
Metab Brain Dis ; 37(7): 2467-2481, 2022 10.
Article in English | MEDLINE | ID: mdl-35867181

ABSTRACT

Development of neuropsychiatric disorder is associated with stress-related increase in pro-inflammatory cytokines. Chrysophyllum albidum fruit is an edible tropical fruit containing vitamins and phenolic compounds, well known for their anti-inflammatory and antioxidant activities. This study was designed to investigate the neuroprotective effect of C. albidum fruit extract (CAFE) on stress and lipopolysaccharide (LPS)-induced behavioral and neurochemical impairments in mice. Male Swiss mice were divided into 6 groups (n = 6). Groups 1-3 were orally treated daily for 14 days with normal saline (0.1 mL/10 g), CAFE (100 mg/kg) and Ferulic acid (FA, 10 mg/kg), and left in home cage as controls. Groups 4-6 were treated similarly but subjected to repeated social defeat (RSD) stress using the resident-intruder model from days 1-14. The RSD-animals were injected with LPS (125 µg/kg, i.p) 60 min after each RSD session from days 8-14. Neurobehavioral functions: locomotor, cognitive and anxiety-like behaviors were assessed 24 h after the last treatment. Pro-inflammatory cytokines (IL-1ß, IL-6 and TNF-α), dopamine, acetylcholinesterase, glutamic acid decarboxylase (GAD), malondialdehyde, nitrites, and reduced glutathione (GSH) were determined in brain tissue. CAFE significantly attenuated RSD and LPS-induced hypolocomotion, cognitive impairment and anxiety-like behavior when compared to the control. Treatment with CAFE also significantly reversed the negative effects of RSD and LPS on pro-inflammatory cytokines, dopamine, acetylcholinesterase, GAD, and oxidative-nitrosative stress levels. The findings clearly indicated that Chrysophyllum albidum fruit demonstrated neuroprotective effects and can play a key role in mitigating against chronic stress and inflammation linked to neuropsychiatric disorders.


Subject(s)
Neuroprotective Agents , Sapotaceae , Animals , Mice , Male , Antioxidants/pharmacology , Antioxidants/therapeutic use , Lipopolysaccharides/pharmacology , Acetylcholinesterase , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Social Defeat , Fruit/chemistry , Fruit/metabolism , Tumor Necrosis Factor-alpha/metabolism , Interleukin-6 , Nitrites/analysis , Nitrites/pharmacology , Dopamine , Glutamate Decarboxylase/analysis , Glutamate Decarboxylase/pharmacology , Saline Solution/pharmacology , Sapotaceae/chemistry , Sapotaceae/metabolism , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Glutathione/pharmacology , Cytokines , Malondialdehyde/pharmacology , Vitamins , Oxidative Stress
3.
Inflammopharmacology ; 30(5): 1891-1907, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35876932

ABSTRACT

Schizophrenia is a common mental disorder affecting patients' thoughts, behavior, and cognition. Recently, the NRG1/ErbB4 signaling pathway emerged as a candidate therapeutic target for schizophrenia. This study investigates the effects of aripiprazole and sertindole on the NRG1/ErbB4 and PI3K/AKT/mTOR signaling pathways in ketamine-induced schizophrenia in rats. Young male Wistar rats received ketamine (30 mg/kg, intraperitoneally) for 5 consecutive days and aripiprazole (3 mg/kg, orally) or sertindole (2.5 mg/kg, orally) for 14 days. The proposed pathway was investigated by injecting LY294002 (a selective PI3K inhibitor) (25 µg/kg, intrahippocampal injection) 30 min before the drugs. Twenty-four hours after the last injection, animals were subjected to behavioral tests: the open field test, sucrose preference test, novel object recognition task, and social interaction test. Both aripiprazole and sertindole significantly ameliorated ketamine-induced schizophrenic-like behavior, as expected, because of their previously demonstrated antipsychotic activity. Besides, both drugs alleviated ketamine-induced oxidative stress and neurotransmitter level changes in the hippocampus. They also increased the gamma-aminobutyric acid and glutamate levels and glutamate decarboxylase 67 and parvalbumin mRNA expression in the hippocampus. Moreover, aripiprazole and sertindole increased the NRG1 and ErbB4 mRNA expression levels and PI3K, p-Akt, and mTOR protein expression levels. Interestingly, pre-injecting LY294002 abolished all the effects of the drugs. This study reveals that the antipsychotic effects of aripiprazole and sertindole are partly due to oxidative stress reduction as well as NRG1/ErbB4 and PI3K/AKT/mTOR signaling pathways activation. The NRG1/ErbB4 and PI3K signaling pathways may offer a new therapeutic approach for treating schizophrenia in humans.


Subject(s)
Antipsychotic Agents , Ketamine , Schizophrenia , Animals , Antipsychotic Agents/adverse effects , Aripiprazole/adverse effects , Glutamate Decarboxylase/metabolism , Glutamate Decarboxylase/pharmacology , Glutamate Decarboxylase/therapeutic use , Glutamates/adverse effects , Humans , Imidazoles , Indoles , Ketamine/pharmacology , Male , Neuregulin-1/genetics , Neuregulin-1/metabolism , Neuregulin-1/pharmacology , Parvalbumins/adverse effects , Parvalbumins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor, ErbB-4/genetics , Receptor, ErbB-4/metabolism , Receptor, ErbB-4/therapeutic use , Schizophrenia/chemically induced , Schizophrenia/drug therapy , Signal Transduction , Sucrose/adverse effects , TOR Serine-Threonine Kinases/metabolism , gamma-Aminobutyric Acid
4.
Cerebellum ; 19(5): 715-721, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32592031

ABSTRACT

A prompt diagnosis and treatment of patients with autoimmune cerebellar ataxia (CA) with antibodies against glutamic acid decarboxylase (GAD-Abs) may lead to a better prognosis. Herein, we report prodromal transient neurological symptoms that should raise clinical suspicion of CA with GAD-Abs. We initially identified a 70-year-old man who presented a first acute episode of vertigo, diplopia, and ataxia lasting 2 weeks. Two months later, he experienced a similar episode along with new-onset gaze-evoked nystagmus. After 4 months, downbeat nystagmus, left limb dysmetria, and gait ataxia progressively appeared, and an autoimmune CA was diagnosed based on the positivity of GAD-Abs in serum and cerebrospinal fluid (CSF). We searched retrospectively for similar presentations in a cohort of 31 patients diagnosed with CA and GAD-Abs. We found 11 (35.4%) patients (all women, median age 62 years; 8/11 [72.7%] with autoimmune comorbidities) with transient neurological symptoms antedating CA onset by a median of 3 months, including vertigo in 9 (81.8%; described as paroxysmal in 8) and fluctuating diplopia in 3 (27.3%) patients. The identification of transient neurological symptoms of unknown etiology, such as paroxysmal vertigo and fluctuating diplopia, should lead to GAD-Abs testing in serum and CSF, especially in patients with autoimmune comorbidities.


Subject(s)
Cerebellar Ataxia/drug therapy , Gait Ataxia/drug therapy , Glutamate Decarboxylase/pharmacology , Stiff-Person Syndrome/drug therapy , Aged , Autoantibodies/blood , Cerebellar Ataxia/complications , Cerebellar Ataxia/diagnosis , Glutamate Decarboxylase/immunology , Humans , Retrospective Studies , Stiff-Person Syndrome/complications
5.
Clin Exp Immunol ; 197(3): 329-340, 2019 09.
Article in English | MEDLINE | ID: mdl-31009057

ABSTRACT

Type 1 diabetes mellitus is an autoimmune metabolic disorder characterized by chronic hyperglycemia, the presence of autoreactive T and B cells and autoantibodies against self-antigens. A membrane-bound enzyme on the pancreatic beta-cells, glutamic acid decarboxylase 65 (GAD65), is one of the main autoantigens in type 1 diabetes. Autoantibodies against GAD65 are potentially involved in beta-cell destruction and decline of pancreatic functions. The human complement receptor type 1 (CD35) on B and T lymphocytes has a suppressive activity on these cells. We hypothesized that it may be possible to eliminate GAD65-specific B cells from type 1 diabetes patients by using chimeric molecules, containing an anti-CD35 antibody, coupled to peptides resembling GAD65 B/T epitopes. These molecules are expected to selectively bind the anti-GAD65 specific B cells by the co-cross-linking of the immunoglobulin receptor and CD35 and to deliver a suppressive signal. Two synthetic peptides derived from GAD65 protein (GAD65 epitopes) and anti-CD35 monoclonal antibody were used for the construction of two chimeras. The immunomodulatory activity of the engineered antibodies was tested in vitro using peripheral blood mononuclear cells (PBMCs) from type 1 diabetes patients. A reduction in the number of anti-GAD65 IgG antibody-secreting plasma cells and increased percentage of apoptotic B lymphocytes was observed after treatment of these PBMCs with the engineered antibodies. The constructed chimeric molecules are able to selectively modulate the activity of GAD65-specific B lymphocytes and the production of anti-GAD65 IgG autoantibodies by co-cross-linking of the inhibitory CD35 and the B cell antigen receptor (BCR). This treatment presents a possible way to alter the autoimmune nature of these cells.


Subject(s)
Antibodies, Monoclonal , Epitopes, B-Lymphocyte , Glutamate Decarboxylase , Peptides , Protein Engineering , Receptors, Complement 3b , Adult , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacology , Autoantibodies/genetics , Autoantibodies/immunology , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/pharmacology , Female , Glutamate Decarboxylase/chemistry , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/pharmacology , Humans , Male , Peptides/chemistry , Peptides/genetics , Peptides/pharmacology , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology , Receptors, Complement 3b/antagonists & inhibitors , Receptors, Complement 3b/genetics , Receptors, Complement 3b/immunology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology
6.
J Diabetes Res ; 2018: 9391845, 2018.
Article in English | MEDLINE | ID: mdl-30009185

ABSTRACT

GAD-alum given into lymph nodes to type 1 diabetes patients participating in an open-label pilot trial resulted in preservation of C-peptide similar to promising results from other trials. Here, we compared the immunomodulatory effect of giving GAD-alum directly into lymph nodes versus that induced by subcutaneous administration. Samples from T1D patients (n = 6) who received 4 µg GAD-alum into lymph nodes (LNs), followed by two booster injections one month apart, and from patients (n = 6) who received two subcutaneous injections (SC) (20 µg) given one month apart were compared. GADA, IA-2A, GADA subclasses, IgE, GAD65-induced cytokines, PBMC proliferation, and T cell markers were analyzed. Lower doses of GAD-alum into LN induced higher GADA levels than SC injections and reduced proliferation and IgG1 GADA subclass, while enhancing IgG2, IgG3, and IgG4. The cytokine profile was dominated by the Th2-associated cytokine IL-13, and GAD65 stimulation induced activated CD4 T cells. Patients responding clinically best account for most of the immunological changes. In contrast, SC treatment resulted in predominant IgG1, predominant IFN-γ, higher proliferation, and activated CD4 and CD8 cells. Patients from the LN group with best metabolic outcome seemed to have common immune correlates related to the treatment. This trial is registered with DIAGNODE (NCT02352974, clinicaltrials.gov) and DIABGAD (NCT01785108, clinicaltrials.gov).


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Glutamate Decarboxylase/pharmacology , Th2 Cells/immunology , Adolescent , Alum Compounds/pharmacology , Cell Proliferation , Child , Cytokines/immunology , Female , Humans , Immunoglobulin G/immunology , Immunomodulation , Immunophenotyping , Leukocytes, Mononuclear/immunology , Lymph Nodes/immunology , Lymphocyte Activation , Male , Pilot Projects , Treatment Outcome , Young Adult
7.
Indian J Med Res ; 145(6): 767-776, 2017 Jun.
Article in English | MEDLINE | ID: mdl-29067979

ABSTRACT

BACKGROUND & OBJECTIVES: Type-1 diabetes mellitus (T1DM) and latent autoimmune diabetes in adults (LADA) share similar pathological features but differ in age of onset and progression. There is a scarcity of information on differences in CD4+ T-cell responses, particularly, cytokine secretion, between the two forms of autoimmune diabetes. Here proliferative potential and concentration of pro- and anti-inflammatory cytokines secreted by peripheral blood mononuclear cells (PBMCs) of T1DM and LADA patients were compared, after in vitro stimulation with ß-cell autoantigens. METHODS: A total of 19 patients with LADA, 37 with T1DM and 20 healthy controls were compared on the basis of lymphocyte proliferation and secretion of pro- and anti-inflammatory cytokines belonging to different T-helper types after in vitro stimulation of PBMCs with insulin and glutamic acid decarboxylase 65 (GAD65). RESULTS: Following insulin stimulation, LADA group secreted higher concentration of interleukin-17 (IL-17) (P=0.02) and had higher proportion of interferon gamma (IFN-γ) secretors (P<0.001) than T1DM group. Post-GAD65 stimulation, higher proportion of LADA patients secreted IL-23 than T1DM group (P=0.02). Proportion of responders , as well as levels of secreted IL-10, were significantly higher in LADA than T1DM group, following stimulation with both insulin (P=0.01) and GAD65 (P=0.03). A significant positive correlation was observed between body mass index and IL-17 levels (r=0.41, P=0.04) and fasting plasma C-peptide with IL-10 levels (r=0.37, P=0.04). INTERPRETATION & CONCLUSIONS: There are differences in the portfolio of cytokine secretion in diabetic subjects with varying rates of ß-cell destruction as LADA subjects secrete higher levels of both pro- and anti-inflammatory cytokines on exposure to ß-cell autoantigens, thus highlighting another distinguishing feature in the pathophysiology of the two forms of autoimmune diabetes.


Subject(s)
Cytokines/blood , Diabetes Mellitus, Type 1/blood , Diagnosis, Differential , Latent Autoimmune Diabetes in Adults/blood , Adolescent , Adult , Autoantibodies/blood , Autoantigens/blood , CD4-Positive T-Lymphocytes/metabolism , Child , Diabetes Mellitus, Type 1/pathology , Female , Glutamate Decarboxylase/pharmacology , Humans , Insulin/pharmacology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Interferon-gamma/blood , Interleukin-10/blood , Interleukin-17/blood , Interleukin-23/blood , Latent Autoimmune Diabetes in Adults/pathology , Leukocytes, Mononuclear/metabolism , Male , Young Adult
8.
Clin Immunol ; 154(1): 72-83, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24993292

ABSTRACT

Tolerogenic dendritic cells (tDC) constitute a promising therapy for autoimmune diseases, since they can anergize T lymphocytes recognizing self-antigens. Patients with type 1 diabetes mellitus (T1D) have autoreactive T cells against pancreatic islet antigens (insulin, glutamic acid decarboxylase 65 -GAD65-). We aimed to determine the ability of tDC derived from T1D patients to inactivate their insulin- and GAD65-reactive T cells. CD14+ monocytes and CD4+CD45RA- effector/memory lymphocytes were isolated from 25 patients. Monocyte-derived DC were generated in the absence (control, cDC) or presence of IL-10 and TGF-ß1 (tDC), and loaded with insulin or GAD65. DC were cultured with T lymphocytes (primary culture), and cell proliferation and cytokine secretion were determined. These lymphocytes were rechallenged with insulin-, GAD65- or candidin-pulsed cDC (secondary culture) to assess whether tDC rendered T cells hyporesponsive to further stimulation. In the primary cultures, tDC induced significant lower lymphocyte proliferation and IL-2 and IFN-γ secretion than cDC; in contrast, tDC induced higher IL-10 production. Lymphocytes from 60% of patients proliferated specifically against insulin or GAD65 (group 1), whereas 40% did not (group 2). Most patients from group 1 had controlled glycemia. The secondary cultures showed tolerance induction to insulin or GAD65 in 14 and 10 patients, respectively. A high percentage of these patients (70-80%) belonged to group 1. Importantly, tDC induced antigen-specific T-cell hyporesponsiveness, since the responses against unrelated antigens were unaffected. These results suggest that tDC therapy against multiple antigens might be useful in a subset of T1D patients.


Subject(s)
Dendritic Cells/drug effects , Dendritic Cells/immunology , Diabetes Mellitus, Type 1/immunology , Glutamate Decarboxylase/pharmacology , Insulin/pharmacology , Peptide Fragments/pharmacology , T-Lymphocytes/drug effects , Adolescent , Adult , Autoantigens/drug effects , Biological Assay , Cell Proliferation/drug effects , Cells, Cultured , Child , Diabetes Mellitus, Type 1/pathology , Female , Flow Cytometry , Humans , Immune Tolerance , In Vitro Techniques , Male , Middle Aged , T-Lymphocytes/immunology
10.
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
11.
Diabet Med ; 29(10): 1272-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22587593

ABSTRACT

AIM: The balance between T helper cell subsets is an important regulator of the immune system and is often examined after immune therapies. We aimed to study the immunomodulatory effect of glutamic acid decarboxylase (GAD) 65 formulated with aluminium hydroxide (GAD-alum) in children with Type 1 diabetes, focusing on chemokines and their receptors. METHODS: Blood samples were collected from 70 children with Type 1 diabetes included in a phase II clinical trial with GAD-alum. Expression of CC chemokine receptor 5 (CCR5) and CCR4 was analysed on CD4+ and CD8+ lymphocytes after in vitro stimulation with GAD(65) using flow cytometry, and secretion of the chemokines CCL2, CCL3 and CCL4 was detected in peripheral blood mononuclear cell supernatants with Luminex. RESULTS: Expression of Th1-associated CCR5 was down-regulated following antigen challenge, together with an increased CCR4/CCR5 ratio and CCL2 secretion in GAD-alum-treated patients, but not in the placebo group. CONCLUSION: Our results suggest that GAD-alum treatment has induced a favourable immune modulation associated with decreased Th1/Tc1 phenotypes upon antigen re-challenge, which may be of importance for regulating GAD(65) immunity.


Subject(s)
Aluminum Hydroxide/therapeutic use , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Diabetes Mellitus, Type 1/drug therapy , Glutamate Decarboxylase/therapeutic use , T-Lymphocytes, Cytotoxic/drug effects , Th1 Cells/drug effects , Adolescent , Aluminum Hydroxide/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Child , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/immunology , Female , Flow Cytometry , Glutamate Decarboxylase/pharmacology , Humans , Immunologic Memory , Lymphocyte Activation/drug effects , Male , Phenotype , Receptors, CCR4/drug effects , Receptors, CCR5/drug effects , Sweden
12.
PLoS One ; 6(12): e29008, 2011.
Article in English | MEDLINE | ID: mdl-22174945

ABSTRACT

A phase II clinical trial with glutamic acid decarboxylase (GAD) 65 formulated with aluminium hydroxide (GAD-alum) has shown efficacy in preserving residual insulin secretion in children and adolescents with recent-onset type 1 diabetes (T1D). We have performed a 4-year follow-up study of 59 of the original 70 patients to investigate long-term cellular and humoral immune responses after GAD-alum-treatment. Peripheral blood mononuclear cells (PBMC) were stimulated in vitro with GAD(65). Frequencies of naïve, central and effector memory CD4+ and CD8+ T cells were measured, together with cytokine secretion, proliferation, gene expression and serum GAD(65) autoantibody (GADA) levels. We here show that GAD-alum-treated patients display increased memory T-cell frequencies and prompt T-cell activation upon in vitro stimulation with GAD(65), but not with control antigens, compared with placebo subjects. GAD(65)-induced T-cell activation was accompanied by secretion of T helper (Th) 1, Th2 and T regulatory cytokines and by induction of T-cell inhibitory pathways. Moreover, post-treatment serum GADA titres remained persistently increased in the GAD-alum arm, but did not inhibit GAD(65) enzymatic activity. In conclusion, memory T- and B-cell responses persist 4 years after GAD-alum-treatment. In parallel to a GAD(65)-induced T-cell activation, our results show induction of T-cell inhibitory pathways important for regulating the GAD(65) immunity.


Subject(s)
Aluminum Hydroxide/therapeutic use , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , Glutamate Decarboxylase/therapeutic use , Immunity/immunology , Adolescent , Aluminum Hydroxide/blood , Aluminum Hydroxide/pharmacology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Child , Cytokines/metabolism , Diabetes Mellitus, Type 1/blood , Gene Expression Regulation/drug effects , Glutamate Decarboxylase/blood , Glutamate Decarboxylase/pharmacology , Humans , Immunity/drug effects , Immunologic Memory/drug effects , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocyte Count , Real-Time Polymerase Chain Reaction
13.
Diabetes ; 60(6): 1716-25, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21617187

ABSTRACT

OBJECTIVE: Immunotherapy using peptides from the ß-cell antigen GAD65 can preserve glucose homeostasis in diabetes-prone NOD mice; however, the precise mechanisms that arrest islet-reactive T cells remain unresolved. Our previous work revealed that a dominant GAD65 epitope contained two overlapping I-A(g7)-restricted determinants, 524-538 and 530-543, with the former associated with amelioration of hyperglycemia. Here, we sought to discover whether p524-538-specific T cells could directly regulate islet-reactive T cells. RESEARCH DESIGN AND METHODS: Prediabetic NOD mice were used to determine the relationship between peptide p524-538-induced interleukin (IL)-13 and regulation of islet autoimmunity. Pancreatic lymph node (PLN) cells from mice at distinct stages of islet inflammation, peri-insulitis versus invasive insulitis, were harvested to establish the expression pattern of IL-13 receptor α1 (IL-13Rα1) on islet-associated T cells. RESULTS: Peptide p524-538 preferentially induced IL-13-producing T cells that antagonized the release of γ-interferon by spontaneously arising GAD65 autoimmunity, and recombinant human IL-13 inhibited proliferation of islet-reactive clonotypic T cells. A subset of CD4(+) T cells in NOD and NOD.BDC2.5 T cell receptor transgenic mice expressed functional IL-13Rα1, which induced phosphorylation of signal transducer and activator of transcription 6 in the presence of cognate cytokine. Notably, the number of IL-13Rα1(+) T cells was heightened in the PLN of young NOD mice when compared with older female counterparts with advanced insulitis. Immunization with p524-538 preserved IL-13Rα1 expression on PLN T cells. CONCLUSIONS: IL-13 may be important for regulating autoimmunity in the early stages of insulitis, and the loss of IL-13Rα1 on islet-reactive T cells may be a biomarker for fading regional immune regulation and progression to overt diabetes.


Subject(s)
Insulin-Secreting Cells/metabolism , Interleukin-13 Receptor alpha1 Subunit/metabolism , Animals , Autoimmunity/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cell Line , Cell Proliferation/drug effects , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Female , Flow Cytometry , Glutamate Decarboxylase/pharmacology , Humans , Insulin-Secreting Cells/drug effects , Interferon-gamma/metabolism , Interleukin-13/metabolism , Interleukin-13 Receptor alpha1 Subunit/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Peptide Fragments/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , STAT6 Transcription Factor/metabolism
14.
J Clin Endocrinol Metab ; 95(8): 3788-97, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20466784

ABSTRACT

CONTEXT: Mesenchymal stem cells (MSCs) exert an immunosuppressive effect on the immune system. However, studies on the immunomodulatory potential of MSCs in type 1 diabetes are lacking. OBJECTIVE: We aimed to evaluate whether human MSCs may inhibit in vitro pancreatic islet antigen-specific T cell activation in type 1 diabetes. DESIGN: Human MSCs were isolated and characterized. Peripheral blood mononuclear cells (PBMCs) were obtained from nine type 1 diabetic patients at disease onset and 13 healthy control subjects. IFN-gamma, IL-10, and IL-4 enzyme-linked immunospot responses of lymphocytes incubated with glutamic acid decarboxylase 65 (GAD65) were investigated in PBMC cultures and PBMC/MSC cocultures. Levels of prostaglandin E2 (PGE2), IFN-gamma, IL-4, and IL-10 in supernatants were measured by ELISA. PGE2 inhibition experiments with NS-398 and indomethacin were also performed. RESULTS: Five diabetic patients were identified with a positive PBMC IFN-gamma response to GAD65 and negative IL-10 and IL-4 response. PBMC/MSC cocultures resulted in a significant decrease in the number of spots and in detection of IL-4-secreting cells. PGE2 inhibitors abrogated the immune-suppressive effect, indicating an involvement of PGE2 production, and the constitutive production of PGE2 by MSCs was enhanced in PBMC/MSC coculture. Moreover, in GAD-responder patients, GAD-stimulated PBMC/MSC cocultures significantly decreased secretion of IFN-gamma and IL-10 and increased secretion of IL-4. CONCLUSIONS: These results provide evidence that human MSCs abrogate in vitro a proinflammatory T helper type 1 response to an islet antigenic stimulus in type 1 diabetes. MSCs induce IL-4-producing cells, suggesting a possible switch to an antiinflammatory T helper type 2 signaling of T cells.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Glutamate Decarboxylase/pharmacology , Immunity, Cellular/immunology , Mesenchymal Stem Cells/immunology , T-Lymphocytes/immunology , Cells, Cultured , Coculture Techniques , Diabetes Mellitus, Type 1/metabolism , Dinoprostone/immunology , Dinoprostone/metabolism , Enzyme-Linked Immunosorbent Assay , Glutamate Decarboxylase/immunology , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-4/immunology , Interleukin-4/metabolism , Islets of Langerhans/immunology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mesenchymal Stem Cells/cytology , Statistics, Nonparametric , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
15.
Scand J Immunol ; 70(2): 149-58, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19630921

ABSTRACT

UNLABELLED: Perfect maternal diabetes compensation is crucial for the outcome of the baby. However, little is known how hyperglycaemia influences the specific immune response. Furthermore, babies of type 1 diabetes (T1D) mothers have less risk of development T1D than babies with a T1D father. This study aimed to analyze the effect of maternal hyperglycaemia on newborns with focus on the response to diabetes-associated autoantigens. POPULATIONS: (1) Newborns of T1D mothers split into groups according to maternal diabetes compensation during the 3rd trimester: perfect (n = 15) or acceptable (n = 25) compensation. (2) newborns with T1D father (n = 12) (3) newborns with a mother treated for either gestational or type 2 diabetes (n = 10) (4) control newborns (n = 25). Spontaneous as well as diabetes-associated autoantigen-stimulated production of 23 cytokines and chemokines were tested using protein microarray. In addition, the influence of glucose on cytokine and chemokine responsiveness was analyzed in vitro. The study groups differed in their spontaneous as well as stimulated cytokine and chemokine spectra. A prominent Th1 response (high IFN-gamma) from autoantigen stimulation was observed especially in babies of T1D fathers (P = 0.001) and also in mothers with perfect diabetes compensation during the 3rd trimester (P = 0.016) in comparison with control newborns. By contrast, cord blood mononuclear cells cultivated in vitro in high glucose concentration decreased the diabetogenic stimulated Th1 cytokine response. Maternal 'sweet' as well as 'autoimmune environment' may both lead to lower occurrence of T1D within their offspring. Further studies will reveal the exact immunological mechanism of this observation.


Subject(s)
Autoantigens/immunology , Diabetes Mellitus, Type 1/immunology , Fetal Blood/immunology , Hyperglycemia/immunology , Leukocytes, Mononuclear/immunology , Pregnancy in Diabetics/immunology , Adult , Cytokines/biosynthesis , Cytokines/drug effects , Cytokines/immunology , Female , Fetal Blood/metabolism , Glucose/pharmacology , Glutamate Decarboxylase/pharmacology , Humans , Infant, Newborn , Leukocytes, Mononuclear/drug effects , Pregnancy , Protein Array Analysis
16.
Clin Exp Immunol ; 153(2): 174-81, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18549445

ABSTRACT

Extracorporeal photochemotherapy (ECP) has demonstrated immunological effects. The proposed cytotoxic lymphocyte antigen 4 (CTLA-4) involvement, together with forkhead box P3 (FoxP3) and transforming growth factor (TGF)-beta are associated with regulatory T cell activity. The aim of the study was to evaluate the regulatory T cell-associated effect of ECP in recent onset type 1 diabetic (T1D) children. Children (n = 20) with T1D received photopheresis 8-methoxypsoralen + ECP or placebo + shampheresis. Peripheral blood mononuclear cells (PBMC) collected pretreatment (day 1) and post-treatment (day 90) were stimulated with phytohaemagglutinin (PHA) and T1D-associated glutamic acid decarboxylase 65 (GAD(65)) peptide a.a. 247-279. CTLA-4, sCTLA-4, FoxP3 and TGF-beta mRNA transcription was quantified. Photopheresis-treated individuals' relative mRNA expression was generally maintained during the course of the study. Placebo individuals increased in spontaneous CTLA-4 mRNA (P < 0.05) but decreased in expression after stimulation with GAD(65)-peptide (P < 0.05) and PHA (P < 0.05). Spontaneous TGF-beta (P < 0.05) increased whereas PHA- (P < 0.01) and GAD(65)-peptide (P < 0.01)-induced TGF-beta expression decreased in the placebo group, whereas it was maintained in the treated group. Without intervention, expression of CTLA-4 and TGF-beta, stimulated with PHA and GAD(65) peptide, decreased with time, with a parallel reduction of GAD(65)-peptide and PHA-stimulated TGF-beta expression. These parameters were counteracted by ECP. In conclusion, our results indicate that ECP maintains regulatory T cell-associated activity in recent-onset T1D.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Photopheresis , T-Lymphocytes, Regulatory/immunology , Adolescent , Antigens, CD/blood , Antigens, CD/genetics , Biomarkers , CTLA-4 Antigen , Case-Control Studies , Child , Female , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Glutamate Decarboxylase/pharmacology , Humans , Immune Tolerance , Lymphocyte Activation , Male , Phytohemagglutinins/pharmacology , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction/methods , Statistics, Nonparametric , Transforming Growth Factor beta/genetics
17.
J Neurosci Res ; 85(4): 855-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17253644

ABSTRACT

Although the majority of agents with antiexcitotoxic action act as glutamate receptor antagonists, enzymatic degradation of glutamate can also be neuroprotective. The very low specific activity of the mammalian form of glutamate decarboxylase (GAD), the enzyme that catalyzes the formation of gamma-aminobutyric acid (GABA) from glutamate in neurons, is likely to limit its utility as an antiglutamate neuroprotectant. In contrast, the bacterial form of GAD can be isolated with relatively high specific activity and is most active in acidic environments. We have expressed and purified GAD from Escherichia coli (bGAD) and tested the ability of the enzyme to protect against glutamate excitotoxicity. Incubation of rat hipppocampal slices with the potassium channel antagonist tetraethyl ammonium (TEA) resulted in widespread excitotoxic death of pyramidal and granule cell neurons. bGAD alone showed no significant neurotoxicity and significantly reduced excitotoxicity induced by TEA. We hypothesize that bGAD may be internalized into the synaptic vesicle compartment by nonspecific endocytosis, where both the appropriate pH and high glutamate concentrations are present. Targeting of this enzyme to the interior of synaptic vesicles may enhance its potency as a neuroprotectant against excitotoxicity.


Subject(s)
Glutamate Decarboxylase/pharmacology , Hippocampus/injuries , Hippocampus/pathology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Animals, Newborn , Bacterial Proteins/pharmacology , Cell Death/drug effects , Dose-Response Relationship, Drug , Neurons/cytology , Organ Culture Techniques , Potassium Channel Blockers/toxicity , Rats , Rats, Sprague-Dawley , Tetraethylammonium/toxicity
18.
Spine (Phila Pa 1976) ; 31(14): 1555-8, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16778687

ABSTRACT

STUDY DESIGN: A prospective in vivo animal study. OBJECTIVES: To examine the effect of the proenkephalin and glutamic acid decarboxylase (GAD)-expressing herpes simplex virus (HSV)-based vectors in a rodent model of lumbar radiculopathy. SUMMARY OF BACKGROUND DATA: We have previously shown that nonreplicating HSV-based vectors coding for proenkephalin or GAD can be used to transduce dorsal root ganglion (DRG) neurons in vivo to produce enkephalin or gamma-aminobutyric acid. HSV-mediated gene transfer of proenkephalin or GAD to DRG reduces pain-related behavior in rodent models of peripheral neuropathic pain. METHODS: We created a model of lumbar radiculopathy by ligation of the dorsal and ventral lumbar roots proximal to the DRG. Three days later, we inoculated nonreplicating HSV-based vectors coding for proenkephalin or GAD subcutaneously in the foot. RESULTS: Subcutaneous inoculation of either vector 3 days after ligation of the dorsal and ventral L5 lumbar roots resulted in a substantial and significant reduction in pain-related behavior (mechanical allodynia). Vector-mediated reduction in pain-related behavior was higher in magnitude and longer in duration after inoculation of the GAD-expressing vector than that achieved by the inoculation of the proenkephalin-expressing vector. CONCLUSIONS: HSV-mediated gene transfer provides a novel method for treating chronic neuropathic pain related to lumbar root injury in rodents.


Subject(s)
Enkephalins/genetics , Gene Transfer Techniques , Glutamate Decarboxylase/genetics , Pain/physiopathology , Protein Precursors/genetics , Radiculopathy/physiopathology , Animals , Enkephalins/pharmacology , Foot , Ganglia, Spinal , Genetic Vectors , Glutamate Decarboxylase/pharmacology , Hyperesthesia/physiopathology , Injections, Subcutaneous , Ligation , Lumbosacral Region , Protein Precursors/pharmacology , Radiculopathy/etiology , Rats , Rats, Sprague-Dawley , Simplexvirus/genetics , Spinal Nerve Roots
19.
J Interferon Cytokine Res ; 26(4): 207-13, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16704296

ABSTRACT

Type 1 diabetes (T1D) is an autoimmune disease suggested to be of a T helper (Th)1-like origin. This study aimed to investigate the Th1-like and Th2-like profile in high-risk individuals during the prediabetic phase and the immunologic effect of treatment with nicotinamide. High-risk first-degree relatives of T1D patients participating in the European Nicotinamide Diabetes Intervention Trial (ENDIT) were treated with either nicotinamide or placebo. Peripheral blood mononuclear cells (PBMC) were obtained during the prediabetic phase and close to the onset of manifest T1D and from nondiabetic high-risk individuals. Using the sensitive enzyme-linked immunospot (ELISPOT) technique to distinguish Th1-like from Th2-like lymphocytes, secretion of interferon-gamma (IFN-gamma) and interleukin-4 (IL-4) was analyzed from PBMCs spontaneously and after in vitro stimulation with the diabetes-associated autoantigens, glutamic acid decarboxylase 65 (GAD65, protein and peptide, aa 247-279), recombinant tyrosine phosphatase (IA-2), and heat shock protein (HSP, aa 437-460). High-risk individuals showed high spontaneous as well as autoantigen-induced IFN-gamma secretion. Secretion of IFN-gamma and the IFN-gamma/IL-4 ratio, induced by autoantigens, decreased in individuals developing T1D (p < 0.05), whereas nondiabetic individuals showed an increased IL-4 response (p < 0.05). Thus, a Th1-dominated cytokine profile observed in high-risk individuals inclined toward a diagnosis of diabetes. Nicotinamide caused decreased spontaneous (p = 0.05) and in vitro autoantigen-induced IFN-gamma secretion (p < 0.05) and may play a role in immune regulation, even though it has not been shown to prevent T1D.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Interferon-gamma/metabolism , Niacinamide/pharmacology , Prediabetic State/immunology , Th1 Cells/immunology , Adolescent , Adult , Autoantigens/pharmacology , Child , Down-Regulation , Glutamate Decarboxylase/pharmacology , Humans , Interleukin-4/metabolism , Isoenzymes/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Male , Niacinamide/therapeutic use , Risk , Th1 Cells/drug effects
20.
Hum Immunol ; 67(4-5): 264-73, 2006.
Article in English | MEDLINE | ID: mdl-16720206

ABSTRACT

Clinical intervention trials evaluating the efficacy of antibody immunotherapy in type 1 diabetes are in progress. We tested effects on prediabetic islet antigen-specific autoreactive T cells of antithymocyte globulin (ATG) and humanized monoclonal antibodies against CD3 (ChAglyCD3) or CD25 (daclizumab) with regard to downmodulation of the target protein, proliferation, cytokine production, complement-dependent cytotoxicity (CDC), antibody-dependent cell cytotoxicity (ADCC), and survival. ATG leads to depletion of autoreactive CD4+ T cells by ADCC, CDC, and apoptosis, whereas anti-CD3 and anti-CD25 inhibited T-cell autoreactivity in a nondepleting fashion. ATG treatment led to a cytokine burst of Th1- and Th2-associated cytokines. Modulation of cytokine release through humanized monoclonal antibodies was moderate and selective: anti-CD25 led to increased release of IL-2 and reduced production of TNFalpha, whereas anti-CD3 decreased release of interferon-y and IL-5 and increased secretion of IL-10. ATG and the humanized monoclonal antibodies displayed contrasting mechanisms of action.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antilymphocyte Serum/pharmacology , Diabetes Mellitus, Type 1/therapy , Immunization, Passive , Immunoglobulin G/pharmacology , T-Lymphocytes/drug effects , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antilymphocyte Serum/therapeutic use , Apoptosis , CD3 Complex/immunology , Cell Proliferation/drug effects , Clone Cells/immunology , Cytokines/metabolism , Daclizumab , Glutamate Decarboxylase/pharmacology , Humans , Immunoglobulin G/therapeutic use , Isoenzymes/pharmacology , Lymphocyte Activation , T-Lymphocytes/cytology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...