Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 184(3): 840-843, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33545037

ABSTRACT

We have recently identified a novel lymphocyte that is a dual expresser (DE) of TCRαß and BCR. DEs in T1D patients are predominated by a public BCR clonotype (clone-x) that encodes a potent autoantigen that cross-activates insulin-reactive T cells. Betts and colleagues were able to detect DEs but alleged to not detect high DE frequency, clone-x, or similar clones in T1D patients. Unfortunately, the authors did not follow our methods and when they did, their flow cytometric data at two sites were conflicting. Moreover, contrary to their claim, we identified clones similar to clone-x in their data along with clones bearing the core motif (DTAMVYYFDYW). Additionally, their report of no increased usage of clone-x VH/DH genes by bulk B cells confirms rather than challenges our results. Finally, the authors failed to provide data verifying purity of their sorted DEs, making it difficult to draw reliable conclusion of their repertoire analysis. This Matters Arising Response paper addresses the Japp et al. (2021) Matters Arising paper, published concurrently in Cell.


Subject(s)
Diabetes Mellitus, Type 1 , B-Lymphocytes , Clone Cells , Humans , Receptors, Antigen, T-Cell, alpha-beta , T-Lymphocytes
2.
Cell ; 177(6): 1583-1599.e16, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31150624

ABSTRACT

T and B cells are the two known lineages of adaptive immune cells. Here, we describe a previously unknown lymphocyte that is a dual expresser (DE) of TCR and BCR and key lineage markers of both B and T cells. In type 1 diabetes (T1D), DEs are predominated by one clonotype that encodes a potent CD4 T cell autoantigen in its antigen binding site. Molecular dynamics simulations revealed that this peptide has an optimal binding register for diabetogenic HLA-DQ8. In concordance, a synthetic version of the peptide forms stable DQ8 complexes and potently stimulates autoreactive CD4 T cells from T1D patients, but not healthy controls. Moreover, mAbs bearing this clonotype are autoreactive against CD4 T cells and inhibit insulin tetramer binding to CD4 T cells. Thus, compartmentalization of adaptive immune cells into T and B cells is not absolute, and violators of this paradigm are likely key drivers of autoimmune diseases.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Adolescent , Adult , Autoantigens/immunology , Child , Child, Preschool , Diabetes Mellitus, Type 1/metabolism , Epitopes/immunology , Female , HEK293 Cells , HLA-DQ Antigens/immunology , HLA-DQ Antigens/ultrastructure , Humans , Lymphocyte Activation/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Middle Aged , Molecular Dynamics Simulation , Peptides , Protein Binding/immunology
3.
Cell Immunol ; 339: 33-40, 2019 05.
Article in English | MEDLINE | ID: mdl-30765202

ABSTRACT

Interleukin-17 (IL-17) is a potent proinflammatory cytokine that protects a host against fungal and extracellular bacterial infections. On the other hand, excessive or dysregulated production of IL-17 underlines susceptibility to autoimmune disease. Consequently, blocking IL-17 has become an effective strategy for modulating several autoimmune diseases, including multiple sclerosis (MS), psoriasis, and rheumatoid arthritis (RA). Notably, however, IL-17 blockade remains ineffective or even pathogenic against important autoimmune diseases such as inflammatory bowel disease (IBD). Furthermore, the efficacy of IL-17 blockade against other autoimmune diseases, including type 1 diabetes (T1D) is currently unknown and waiting results of ongoing clinical trials. Coming years will determine whether the efficacy of IL-17 blockade is limited to certain autoimmune diseases or can be expanded to other autoimmune diseases. These efforts include new clinical trials aimed at testing second-generation agents with the goal of increasing the efficiency, spectrum, and ameliorating side effects of IL-17 blockade. Here we briefly review the roles of IL-17 in the pathogenesis of selected autoimmune diseases and provide updates on ongoing and recently completed trials of IL-17 based immunotherapies.


Subject(s)
Interleukin-17/immunology , Animals , Autoimmune Diseases/immunology , Humans , Immunotherapy/methods , Inflammatory Bowel Diseases/immunology , Multiple Sclerosis/immunology
4.
Cytokine ; 74(1): 27-34, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25481648

ABSTRACT

Interleukin-10 (IL-10) is arguably the most potent anti-inflammatory cytokine. It is produced by almost all the innate and adaptive immune cells. These cells also serve as its targets, indicating that IL-10 secretion and action is highly regulated and perhaps compartmentalized. Consistent with this notion, various efforts directed at systemic administration of IL-10 to modulate autoimmune diseases (type 1 diabetes, multiple sclerosis, rheumatoid arthritis, psoriasis) have produced conflicting and largely inconsequential effects. On the other hand, IL-10 can promote humoral immune responses, enhancing class II expression on B cells and inducing immunoglobulin (Ig) production. Consequently, the high IL-10 level in systemic lupus erythematosus (SLE) patients is considered pathogenic and its blockade ameliorates the disease. In this perspective, we review preclinical findings and results of recent clinical studies using exogenous IL-10 to treat the aforementioned autoimmune diseases. In addition, given the limited success of IL-10 supplementation, we suggest that future studies should be expanded beyond modulating the delivery modes to include developing new strategies to protect and replenish the endogenous sources of IL-10. As an example, we provide evidence that aberrant Fas-mediated deletion of IL-10-producing B cells subverts the immunoregulatory role of IL-10 in autoimmune diabetes and that modulation of the Fas pathway preserves the IL-10-producing B cells and completely protects NOD mice from developing the disease.


Subject(s)
Autoimmune Diseases/therapy , B-Lymphocytes/immunology , Immunotherapy , Interleukin-10/immunology , Interleukin-10/therapeutic use , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/physiopathology , Clinical Trials as Topic , Female , Humans , Interleukin-10/administration & dosage , Interleukin-10/adverse effects , Interleukin-10/biosynthesis , Mice, Inbred NOD , Multiple Sclerosis/immunology , Multiple Sclerosis/physiopathology , Multiple Sclerosis/therapy
5.
Circulation ; 128(8): 785-94, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23857320

ABSTRACT

BACKGROUND: Rosiglitazone improves glycemic control for patients with type 2 diabetes mellitus, but there remains controversy regarding an observed association with cardiovascular hazard. The cardiovascular effects of rosiglitazone for patients with coronary artery disease remain unknown. METHODS AND RESULTS: To examine any association between rosiglitazone use and cardiovascular events among patients with diabetes mellitus and coronary artery disease, we analyzed events among 2368 patients with type 2 diabetes mellitus and coronary artery disease in the Bypass Angioplasty Revascularization Investigation 2 Diabetes (BARI 2D) trial. Total mortality, composite death, myocardial infarction, and stroke, and the individual incidence of death, myocardial infarction, stroke, congestive heart failure, and fractures, were compared during 4.5 years of follow-up among patients treated with rosiglitazone versus patients not receiving a thiazolidinedione by use of Cox proportional hazards and Kaplan-Meier analyses that included propensity matching. After multivariable adjustment, among patients treated with rosiglitazone, mortality was similar (hazard ratio [HR], 0.83; 95% confidence interval [CI], 0.58-1.18), whereas there was a lower incidence of composite death, myocardial infarction, and stroke (HR, 0.72; 95% CI, 0.55-0.93) and stroke (HR, 0.36; 95% CI, 0.16-0.86) and a higher incidence of fractures (HR, 1.62; 95% CI, 1.05-2.51); the incidence of myocardial infarction (HR, 0.77; 95% CI, 0.54-1.10) and congestive heart failure (HR, 1.22; 95% CI, 0.84-1.82) did not differ significantly. Among propensity-matched patients, rates of major ischemic cardiovascular events and congestive heart failure were not significantly different. CONCLUSIONS: Among patients with type 2 diabetes mellitus and coronary artery disease in the BARI 2D trial, neither on-treatment nor propensity-matched analysis supported an association of rosiglitazone treatment with an increase in major ischemic cardiovascular events. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT00006305.


Subject(s)
Angioplasty , Coronary Artery Disease/epidemiology , Coronary Artery Disease/therapy , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/epidemiology , Thiazolidinediones/adverse effects , Thiazolidinediones/therapeutic use , Aged , Comorbidity , Coronary Artery Disease/mortality , Diabetes Mellitus, Type 2/mortality , Female , Follow-Up Studies , Heart Failure/epidemiology , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/therapeutic use , Incidence , Kaplan-Meier Estimate , Male , Middle Aged , Myocardial Infarction/epidemiology , Retrospective Studies , Risk Factors , Rosiglitazone , Stroke/epidemiology , Treatment Outcome
6.
J Diabetes Res ; 2023: 9972132, 2023.
Article in English | MEDLINE | ID: mdl-37589043

ABSTRACT

Background: Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) are potent antihyperglycemic agents with beneficial effects on weight, cardiovascular, and renal outcomes. Physicians lack guidance as to which patients with insulin-requiring type 2 diabetes will respond best to GLP-1 RAs with respect to glycemic control, insulin dose reduction, and weight loss. This study evaluated the efficacy of GLP-1 RAs in patients with type 2 diabetes on insulin and patient factors that may predict a beneficial clinical response. Methods: Adults with type 2 diabetes treated with insulin who had a GLP-1 RA added to their regimen were evaluated retrospectively. Baseline parameters and outcomes at 3, 6, and 12 months were collected. Results: Among the 81 patients included, there was a mean reduction in hemoglobin A1C of 0.94% (SD, 0.26; p = 0.0007), 0.40% (SD, 0.21; p = 0.0636), and 0.58% (SD, 0.23, p = 0.0154) at 3, 6, and 12 months, respectively, following the addition of a GLP-1 RA. There was also a reduction in body weight noted at each time point. Baseline characteristics including BMI, duration of diabetes, and insulin requirement did not significantly affect A1C reduction when GLP-1 RA was added. At 3 months, patients with a random C-peptide that was normal (≥0.8 ng/ml) were significantly more likely to have discontinued insulin than those with random C-peptide that was low (<0.8 ng/ml) (11 of 23 vs. 0 of 7 patients, p = 0.029). Conclusions: The addition of a GLP-1 RA reduced HbA1C, weight, and insulin requirements in this cohort of patients with type 2 diabetes on insulin. BMI, baseline insulin dose, and diabetes duration did not predict response. A C-peptide level ≥ 0.8 ng/ml predicted a beneficial response after 3 months of therapy.


Subject(s)
Diabetes Mellitus, Type 2 , Glucagon-Like Peptide-1 Receptor , Insulin , Adult , Humans , C-Peptide , Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide-1 Receptor/agonists , Glycated Hemoglobin , Insulin/therapeutic use , Retrospective Studies
7.
Clin Case Rep ; 10(3): e05650, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35356161

ABSTRACT

Insulinomas are rare neuroendocrine pancreatic tumors that can be associated with severe episodes of hypoglycemia, leading to significant morbidity and mortality. These tumors are often difficult to localize, and hypoglycemia control can be challenging since glucose levels can be resistant to conventional therapies. Pasireotide is a novel somatostatin analog with a high affinity to multiple somatostatin receptors. It has up to 40 times higher affinity for somatostatin receptor subtype 5 in comparison with octreotide, leading to a higher inhibition of insulin release from beta cells. There are few case reports regarding the use of pasireotide in refractory hyperinsulinemic hypoglycemia. We describe a challenging case of endogenous hyperinsulinemic hypoglycemia refractory to standard medical treatment, in which pasireotide was used. In this case, imaging studies and calcium stimulation testing failed to localize an insulin-secreting tumor in an 83-year-old woman. Glucose levels remained low despite treatment with diazoxide, verapamil, and octreotide, necessitating the use of IV dextrose solutions. After starting subcutaneous (SC) pasireotide 0.9 mg twice a day, there was a significant improvement in the frequency and severity of hypoglycemic events, allowing the patient to be discharged from the hospital without needing IV glucose support.

8.
Cell Rep Med ; 2(12): 100466, 2021 12 21.
Article in English | MEDLINE | ID: mdl-35028608

ABSTRACT

These preliminary data from an ongoing first-in-human phase 1/2, open-label study provide proof-of-concept that pluripotent stem cell-derived pancreatic endoderm cells (PEC-01) engrafted in type 1 diabetes patients become islet cells releasing insulin in a physiologically regulated fashion. In this study of 17 subjects aged 22-57 with type 1 diabetes, PEC-01 cells were implanted subcutaneously in VC-02 macroencapsulation devices, allowing for direct vascularization of the cells. Engraftment and insulin expression were observed in 63% of VC-02 units explanted from subjects at 3-12 months post-implant. Six of 17 subjects (35.3%) demonstrated positive C-peptide as early as 6 months post-implant. Most reported adverse events were related to surgical implant or explant procedures (27.9%) or to side-effects of immunosuppression (33.7%). Initial data suggest that pluripotent stem cells, which can be propagated to the desired biomass and differentiated into pancreatic islet-like tissue, may offer a scalable, renewable alternative to pancreatic islet transplants.


Subject(s)
C-Peptide/metabolism , Cells, Immobilized/cytology , Diabetes Mellitus, Type 1/therapy , Endoderm/cytology , Insulin/metabolism , Pancreas/cytology , Stem Cell Transplantation , Stem Cells/cytology , Adolescent , Adult , Aged , Diabetes Mellitus, Type 1/metabolism , Female , Humans , Male , Middle Aged , Young Adult
10.
World J Diabetes ; 11(1): 13-25, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31938470

ABSTRACT

Type 1 diabetes (T1D) is an autoimmune disease that usually strikes early in life, but can affect individuals at almost any age. It is caused by autoreactive T cells that destroy insulin-producing beta cells in the pancreas. Epidemiological studies estimate a prevalence of 1 in 300 children in the United States with an increasing incidence of 2%-5% annually worldwide. The daily responsibility, clinical management, and vigilance required to maintain blood sugar levels within normal range and avoid acute complications (hypoglycemic episodes and diabetic ketoacidosis) and long term micro- and macro-vascular complications significantly affects quality of life and public health care costs. Given the expansive impact of T1D, research work has accelerated and T1D has been intensively investigated with the focus to better understand, manage and cure this condition. Many advances have been made in the past decades in this regard, but key questions remain as to why certain people develop T1D, but not others, with the glaring example of discordant disease incidence among monozygotic twins. In this review, we discuss the field's current understanding of its pathophysiology and the role of genetics and environment on the development of T1D. We examine the potential implications of these findings with an emphasis on T1D inheritance patterns, twin studies, and disease prevention. Through a better understanding of this process, interventions can be developed to prevent or halt it at early stages.

11.
Br J Psychiatry ; 194(5): 434-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19407273

ABSTRACT

BACKGROUND: Some studies suggest individuals with schizophrenia have an increased risk of diabetes prior to antipsychotic use. Small sample sizes and the potential for confounding by hypercortisolaemia have decreased confidence in those results. AIMS: To examine diabetes-related factors in newly diagnosed, antipsychotic-naive people with non-affective psychosis. METHOD: Participants with psychosis (the psychosis group; n = 50) and matched controls (the control group; n = 50) were given a 2 h oral glucose tolerance test. Fasting concentrations were also determined for adiponectin, interleukin-6 and C-reactive protein. RESULTS: Compared with the control group, the psychosis group had significant increases in 2 h glucose and interleukin-6 concentrations, and in the prevalence of abnormal glucose tolerance (16% of psychosis group v. 0% of control group). Adiponectin and C-reactive protein concentrations did not differ significantly between the two groups. These findings could not be attributed to differences in cortisol concentrations, smoking, gender, neighbourhood of residence, body mass index, aerobic conditioning, ethnicity, socioeconomic status or age. CONCLUSIONS: Individuals with non-affective psychosis appear to have an increased prevalence of abnormal glucose tolerance prior to antipsychotic treatment, as well as abnormalities in a related inflammatory molecule. These underlying problems may contribute to the metabolic side-effects of antipsychotic medications.


Subject(s)
Blood Glucose/metabolism , Glucose Metabolism Disorders/epidemiology , Schizophrenia/metabolism , Adiponectin/blood , Adolescent , Adult , Antipsychotic Agents/therapeutic use , C-Reactive Protein/metabolism , Case-Control Studies , Female , Glucose Metabolism Disorders/metabolism , Glucose Tolerance Test , Humans , Intercellular Signaling Peptides and Proteins/blood , Interleukin-6/blood , Male , Middle Aged , Regression Analysis , Schizophrenia/epidemiology , Young Adult
12.
Schizophr Res ; 98(1-3): 302-6, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18031995

ABSTRACT

INTRODUCTION: We attempted to replicate two previous studies which found an increased risk of diabetes in the relatives of schizophrenia probands. METHODS: N=34 patients with newly-diagnosed nonaffective psychosis and N=52 non-psychiatric controls were interviewed for parental history of Type 2 diabetes. RESULTS: In a logistic regression model that included multiple potential confounders, psychosis was a significant predictor of Type 2 diabetes in either parent (p<0.04). DISCUSSION: We found an increased prevalence of Type 2 diabetes in the parents of nonaffective psychosis subjects. This association may be due to shared environmental or genetic risk factors, or both.


Subject(s)
Child of Impaired Parents , Diabetes Mellitus, Type 2/epidemiology , Family , Parents/psychology , Schizophrenia/epidemiology , Adult , Child , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/genetics , Female , Genetic Predisposition to Disease/genetics , Humans , Logistic Models , Male , Maternal Age , Models, Statistical , Paternal Age , Prevalence , Risk Factors , Schizophrenia/genetics , Schizophrenic Psychology , Social Environment
13.
Med Clin North Am ; 92(2): 407-25, ix-x, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18298986

ABSTRACT

Hyperglycemia is an increasingly common and often complex condition to manage in the inpatient setting. Numerous clinical trials have demonstrated associations between uncontrolled diabetes and poor clinical outcomes in a number of inpatient settings. Insulin remains the treatment of choice for the majority of hyperglycemic hospitalized patients and should be prescribed in a physiologic manner, employing basal and bolus insulin. Intravenous insulin should be used liberally in the ICU setting where randomized studies have demonstrated improved outcomes. Recommendations for insulin use in the inpatient setting are provided.


Subject(s)
Diabetes Mellitus/therapy , Hospitalists/methods , Inpatients , Outcome Assessment, Health Care , Humans , United States
14.
Discov Med ; 26(142): 79-83, 2018 09.
Article in English | MEDLINE | ID: mdl-30399325

ABSTRACT

CD5+ B cells expand in many autoimmune diseases, including type 1 diabetes (T1D), rheumatoid arthritis (RA), and systemic lupus erythematosus (SLE). Furthermore, CD5+ B cells contain important subsets: IL-10-producing B-reg cells, FasL-expressing subset, and the majority of pre-naive B cells. In addition, they are major sources of natural autoantibodies, which are polyreactive and autoreactive. Thus, CD5+ B cells are clearly loaded with autoimmune clues that are yet to be unlocked and understood. We hypothesize that human CD5+ B cells are likely to yield enormously important novel information about the role of B cells in autoimmune disease if analyzed using the new technological advances in molecular biology and genomics. Use of high-throughput sequencing of B cell receptors (BCR) of CD5+ B cells could reveal public BCRs associated with autoimmune diseases, whereas transcriptional analysis of CD5+ B cells using single-cell RNA-seq may delineate distinct sublineages and their relationship to conventional B cells. If it turns out that autoimmune repertoires are concentrated in CD5+ B cells, given that CD5+ B cells are clearly identifiable by flow cytometry, therapeutic strategies can be developed to safely remove CD5+ B cells to mitigate ongoing autoimmunity and protect at-risk individuals.


Subject(s)
Autoimmunity/physiology , B-Lymphocytes/metabolism , CD5 Antigens/metabolism , High-Throughput Screening Assays/methods , Animals , Autoantibodies/genetics , Autoantibodies/immunology , Autoantibodies/metabolism , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmunity/genetics , Humans , Mice , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/metabolism
15.
Front Immunol ; 8: 402, 2017.
Article in English | MEDLINE | ID: mdl-28439273

ABSTRACT

Fas ligand drives insulitis in the non-obese diabetic mouse model of type 1 diabetes (T1D) and negatively regulates IL-10-producing (IL-10pos) CD5+ B cells in pancreata. Relevance of these phenomena to the human disease is poorly understood. Here, using splenocytes from T1D, autoantibody (Ab+), and non-diabetic (ND) human subjects, we show that a subpopulation of CD5+ B cells that is characterized by expression of FasL (FasLhiCD5+) was significantly elevated in T1D subjects, many of whom had significantly reduced frequency of IL-10posCD5+ B cells compared to Ab+ subjects. The majority of FasLhiCD5+ B cells did not produce cytokines and were more highly resistant to activation-induced cell death than their IL-10posCD5+ counterparts. These results associate expansion of FasL-expressing CD5+ B cells with T1D and lay the groundwork for future mechanistic studies to understand specific role in disease pathogenesis.

16.
Discov Med ; 21(115): 213-9, 2016 03.
Article in English | MEDLINE | ID: mdl-27115172

ABSTRACT

Immunotherapy has revolutionized treatment of cancers and autoimmune diseases. Bucking the trend, however, is type 1 diabetes (T1D), although it is one of best understood autoimmune diseases and individuals at genetic risk are identifiable with high certainty. Here we review the major obstacles associated with pan-B-cell-depletion using rituximab (RTX) and discuss the notion that B cell-directed therapy may be most effective as a preventive measure. We suggest that it will be more productive to aim at identifying and targeting autoreactive B cells rather than making adjustments to pan-B cell depletion and that non-conventional alternative therapies such as antibody blockade of FasL to bolster IL-10-producing Breg cells, which work successfully in mice, should be considered.


Subject(s)
B-Lymphocytes/drug effects , Diabetes Mellitus, Type 1/therapy , Immunologic Factors/therapeutic use , Immunotherapy/methods , Rituximab/therapeutic use , Animals , Antigens, CD20/immunology , Antigens, CD20/metabolism , Autoimmunity/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Clinical Trials as Topic , Drug Therapy, Combination , Fas Ligand Protein/antagonists & inhibitors , Fas Ligand Protein/metabolism , Humans , Immunologic Factors/adverse effects , Immunotherapy/adverse effects , Interleukin-10/metabolism , Islets of Langerhans/immunology , Islets of Langerhans Transplantation/immunology , Mice , Rituximab/adverse effects , Treatment Outcome
17.
Vasc Health Risk Manag ; 11: 107-16, 2015.
Article in English | MEDLINE | ID: mdl-25657589

ABSTRACT

Cardiovascular disease is the leading cause of mortality in type 2 diabetes mellitus. Hyperinsulinemia is associated with increased cardiovascular risk, but the effects of exogenous insulin on cardiovascular disease progression have been less well studied. Insulin has been shown to have both cardioprotective and atherosclerosis-promoting effects in laboratory animal studies. Long-term clinical trials using insulin to attain improved diabetes control in younger type 1 and type 2 diabetes patients have shown improved cardiovascular outcomes. Shorter trials of intensive diabetes control with high insulin use in higher risk patients with type 2 diabetes have shown either no cardiovascular benefit or increased all cause and cardiovascular mortality. Glargine insulin is a basal insulin analog widely used to treat patients with type 1 and type 2 diabetes. This review focuses on the effects of glargine on cardiovascular outcomes. Glargine lowers triglycerides, leads to a modest weight gain, causes less hypoglycemia when compared with intermediate-acting insulin, and has a neutral effect on blood pressure. The Outcome Reduction With Initial Glargine Intervention (ORIGIN trial), a 6.2 year dedicated cardiovascular outcomes trial of glargine demonstrated no increased cardiovascular risk.


Subject(s)
Cardiovascular Diseases/prevention & control , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/administration & dosage , Insulin, Long-Acting/administration & dosage , Biomarkers/blood , Blood Glucose/drug effects , Blood Glucose/metabolism , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/mortality , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/mortality , Drug Administration Schedule , Glycated Hemoglobin/metabolism , Humans , Hypoglycemic Agents/adverse effects , Insulin Glargine , Insulin, Long-Acting/adverse effects , Risk Assessment , Risk Factors , Time Factors , Treatment Outcome
18.
Psychiatr Serv ; 55(8): 892-900, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15292539

ABSTRACT

OBJECTIVE: Type 2 diabetes is an important comorbid medical condition associated with schizophrenia. The objective of this study was to compare glycosylated hemoglobin (HbA(1c)) levels of patients who had type 2 diabetes and schizophrenia with those of patients who had type 2 diabetes and major mood disorders and those who had type 2 diabetes but who did not have severe mental illness. METHODS: A sample of 300 patients with type 2 diabetes was recruited from community mental health centers in the greater Baltimore region and nearby primary care clinics. Of these, 100 had schizophrenia, 101 had a major mood disorder, and 99 had no identified severe mental illness. HbA(1c), the main outcome measure, was compared between the group with schizophrenia and the other two groups. RESULTS: All three groups had HbA(1c) values above recommended levels. HbA(1c) levels were significantly lower among patients with schizophrenia than among patients who did not have severe mental illness but were not significantly different from those of patients who had major mood disorders. Patients for whom olanzapine was prescribed had higher HbA(1c) levels than those for whom other antipsychotic agents were prescribed. CONCLUSIONS: All three groups of patients require improved diabetes treatment to achieve acceptable HbA(1c) levels. There may be previously unrecognized benefits for diabetes management among persons with severe mental illnesses who are receiving regular mental heath care, but these individuals may also have risk factors that can influence diabetes outcomes and HbA(1c) levels.


Subject(s)
Benzodiazepines/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/epidemiology , Mental Disorders/drug therapy , Mental Disorders/epidemiology , Body Mass Index , Demography , Diabetes Mellitus, Type 2/physiopathology , Female , Glycated Hemoglobin/metabolism , Humans , Male , Mental Disorders/blood , Middle Aged , Olanzapine , Schizophrenia/blood , Schizophrenia/drug therapy , Schizophrenia/epidemiology , Severity of Illness Index
19.
Diabetes Care ; 37(10): 2746-54, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25011949

ABSTRACT

OBJECTIVE: Previous studies demonstrated that the anti-CD3 monoclonal antibody otelixizumab, administered at a total dose of 48-64 mg, can slow the loss of C-peptide in recent-onset type 1 diabetes patients, with frequent reactivation of Epstein Barr virus (EBV). The DEFEND-1 (Durable Response Therapy Evaluation for Early or New-Onset Type 1 Diabetes) trial was designed to test whether a lower dose of otelixizumab could preserve C-peptide secretion in new-onset type 1 diabetes patients. RESEARCH DESIGN AND METHODS: A multicenter, randomized, placebo-controlled trial was performed in sites in the U.S., Canada, and Europe. Two hundred eighty-one patients were randomized to treatment with 3.1 mg otelixizumab administered over 8 days or placebo. The primary end point of the study was the change in C-peptide area under the curve (AUC) from a 2-h mixed-meal tolerance test at month 12. RESULTS: The change in 2-h C-peptide AUC was not different between placebo-treated patients and otelixizumab-treated patients (-0.20 vs. -0.22 nmol/L, P = 0.81). Secondary end points, including HbA1c, glucose variability, and insulin dose, were also not statistically different between the two groups. More patients in the otelixizumab group than in the placebo group experienced adverse events, mostly grade 1 or grade 2. There was no EBV reactivation (viral load >10,000 copies/10(6) peripheral blood mononuclear cells) in the otelixizumab group, in contrast with previously published studies at higher doses of otelixizumab. CONCLUSIONS: Otelixizumab was well tolerated in patients with recent-onset type 1 diabetes at a total dose of 3.1 mg, but did not achieve preservation of levels of C-peptide or other markers of metabolic control.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/administration & dosage , Adolescent , Adult , Age of Onset , C-Peptide/blood , CD3 Complex/immunology , Canada/epidemiology , Child , Diabetes Mellitus, Type 1/blood , Dose-Response Relationship, Drug , Europe/epidemiology , Female , Humans , Insulin/therapeutic use , Male , Middle Aged , United States , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL