Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Diabetes Res Clin Pract ; 205: 110974, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37884063

ABSTRACT

AIMS: In recent-onset type 1 diabetes, clamp-derived C-peptide predicts good response to anti-CD3. Elevated proinsulin and proinsulin/C-peptide ratio (PI/CP) suggest increased metabolic/inflammatory beta cell burden. We reanalyzed trial data to compare the ability of baseline acutely glucose-stimulated proinsulin, C-peptide and PI/CP to predict functional outcome. METHODS: Eighty recent-onset type 1 diabetes patients participated in the placebo-controlled otelixizumab (GSK; NCT00627146) trial. Hyperglycemic clamps were performed at baseline, 6, 12 and 18 months, involving 3 h of induced euglycemia, followed by acutely raising and maintaining glycemia to ≥ 10 mmol/l for 140 min. Plasma proinsulin, C-peptide and PI/CP were determined after acute (minute 0 at 10 mmol/l; PI0, CP0, PI/CP0) and sustained glucose stimulation (AUC between minutes 60-140). Outcome was assessed as change in AUC60-140 C-peptide from baseline. RESULTS: In multiple linear regression, higher baseline (≥median [P50]) PI0 independently predicted preservation of beta cell function in response to anti-CD3 and interacted significantly with IAA. During follow-up, anti-CD3 tempered a further increase in PI/CP0, but not in PI0. CP0 outperformed PI0 and PI/CP0 for post-treatment monitoring. CONCLUSIONS: In recent-onset type 1 diabetes, elevated acutely glucose-stimulated proinsulin may complement or replace acutely or sustainedly stimulated C-peptide release for identifying good responders to anti-CD3, but not as outcome measure.


Subject(s)
Diabetes Mellitus, Type 1 , Proinsulin , Humans , Proinsulin/metabolism , Proinsulin/therapeutic use , Diabetes Mellitus, Type 1/drug therapy , Insulin/therapeutic use , Glucose/therapeutic use , C-Peptide , Blood Glucose/metabolism
2.
Cell Transplant ; 32: 9636897231167323, 2023.
Article in English | MEDLINE | ID: mdl-37129266

ABSTRACT

Subcutaneous implants of device-encapsulated stem cell-derived pancreatic endoderm (PE) can establish a functional beta cell mass (FBM) with metabolic control in immune-compromised mice. In a study with human-induced pluripotent stem cell-PE, this outcome was favored by a preformed pouch which allowed lesion-free insertion of devices in a pre-vascularized site. This was not reproduced in nude rats, known to exhibit a higher innate reactivity than mice and therefore relevant as preclinical model: a dense fibrotic capsule formed around subcutis (SC) implants with virtually no FBM formation. Placement in omentum (OM) of nude rats provided a less fibrous, better vascularized environment than SC. It resulted in less donor cell loss (56% recovery at post-transplant-PT week 3 versus 16% in SC) allowing FBM-formation. At PT week 30, 6/13 OM-recipients exhibited glucose-induced plasma hu-C-peptide to 0.1-0.4 ng/ml, versus 0/8 in SC-recipients. These levels are more than 10-fold lower than in a state of metabolic control. This shortcoming is not caused by inadequate glucose responsiveness of the beta cells but by their insufficient number. The size of the formed beta cell mass (0.4 ± 0.2 µl) was lower than that reported in mice receiving the same cell product subcutaneously; the difference is attributed to a lower expansion of pancreatic progenitor cells and to their lower degree of differentiation to beta cells. This study in the nude rat model demonstrates that OM provides a better environment for formation of beta cells in device-encapsulated PE-implants than SC. It also identified targets for increasing their dose-efficacy.


Subject(s)
Induced Pluripotent Stem Cells , Insulin-Secreting Cells , Islets of Langerhans Transplantation , Rats , Humans , Mice , Animals , Rats, Nude , Induced Pluripotent Stem Cells/metabolism , Endoderm/metabolism , Omentum , Islets of Langerhans Transplantation/methods , Glucose/metabolism , Cell Differentiation
3.
Cells ; 10(7)2021 07 04.
Article in English | MEDLINE | ID: mdl-34359863

ABSTRACT

Ongoing beta cell death in type 1 diabetes (T1D) can be detected using biomarkers selectively discharged by dying beta cells into plasma. microRNA-375 (miR-375) ranks among the top biomarkers based on studies in animal models and human islet transplantation. Our objective was to identify additional microRNAs that are co-released with miR-375 proportionate to the amount of beta cell destruction. RT-PCR profiling of 733 microRNAs in a discovery cohort of T1D patients 1 h before/after islet transplantation indicated increased plasma levels of 22 microRNAs. Sub-selection for beta cell selectivity resulted in 15 microRNAs that were subjected to double-blinded multicenter analysis. This led to the identification of eight microRNAs that were consistently increased during early graft destruction: besides miR-375, these included miR-132/204/410/200a/429/125b, microRNAs with known function and enrichment in beta cells. Their potential clinical translation was investigated in a third independent cohort of 46 transplant patients by correlating post-transplant microRNA levels to C-peptide levels 2 months later. Only miR-375 and miR-132 had prognostic potential for graft outcome, and none of the newly identified microRNAs outperformed miR-375 in multiple regression. In conclusion, this study reveals multiple beta cell-enriched microRNAs that are co-released with miR-375 and can be used as complementary biomarkers of beta cell death.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Islets of Langerhans Transplantation , MicroRNAs/genetics , Biomarkers/metabolism , Cell Count , Cohort Studies , Gene Expression Profiling , Gene Expression Regulation , Humans , MicroRNAs/metabolism , ROC Curve , Reproducibility of Results , Tropism
4.
Stem Cells Transl Med ; 8(12): 1296-1305, 2019 12.
Article in English | MEDLINE | ID: mdl-31379140

ABSTRACT

Device-encapsulated human stem cell-derived pancreatic endoderm (PE) can generate functional ß-cell implants in the subcutis of mice, which has led to the start of clinical studies in type 1 diabetes. Assessment of the formed functional ß-cell mass (FBM) and its correlation with in vivo metabolic markers can guide clinical translation. We recently reported ex vivo characteristics of device-encapsulated human embryonic stem cell-derived (hES)-PE implants in mice that had established a metabolically adequate FBM during 50-week follow-up. Cell suspensions from retrieved implants indicated a correlation with the number of formed ß cells and their maturation to a functional state comparable to human pancreatic ß cells. Variability in metabolic outcome was attributed to differences in number of PE-generated ß cells. This variability hinders studies on processes involved in FBM-formation. This study reports modifications that reduce variability. It is undertaken with device-encapsulated human induced pluripotent stem cell-derived-PE subcutaneously implanted in mice. Cell mass of each cell type was determined on intact tissue inside the device to obtain more precise data than following isolation and dispersion. Implants in a preformed pouch generated a glucose-controlling ß-cell mass within 20 weeks in over 60% of recipients versus less than 20% in the absence of a pouch, whether the same or threefold higher cell dose had been inserted. In situ analysis of implants indicated a role for pancreatic progenitor cell expansion and endocrine differentiation in achieving the size of ß- and α-cell mass that correlated with in vivo markers of metabolic control. Stem Cells Translational Medicine 2019;8:1296&1305.


Subject(s)
Endoderm/cytology , Glucose/metabolism , Induced Pluripotent Stem Cells/cytology , Insulin-Secreting Cells/cytology , Islets of Langerhans Transplantation/instrumentation , Pancreas/cytology , Animals , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Cells, Cultured , Endoderm/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans Transplantation/methods , Male , Mice , Mice, SCID , Pancreas/metabolism , Tissue Engineering
5.
J Clin Endocrinol Metab ; 104(2): 451-460, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30203041

ABSTRACT

Aim: Several biomarkers have been proposed to detect pancreatic ß cell destruction in vivo but so far have not been compared for sensitivity and significance. Methods: We used islet transplantation as a model to compare plasma concentrations of miR-375, 65-kDa subunit of glutamate decarboxylase (GAD65), and unmethylated insulin DNA, measured at subpicomolar sensitivity, and study their discharge kinetics, power for outcome prediction, and detection of graft loss during follow-up. Results: At 60 minutes after transplantation, GAD65 and miR-375 consistently showed near-equimolar and correlated increases proportional to the number of implanted ß cells. GAD65 and miR-375 showed comparable power to predict poor graft outcome at 2 months, with areas under the curve of 0.833 and 0.771, respectively (P = 0.53). Using receiver operating characteristic analysis, we defined likelihood ratios (LRs) for rationally selected result intervals. In GADA-negative recipients (n = 28), GAD65 <4.5 pmol/L (LR = 0.15) and >12.2 pmol/L (LR = ∞) predicted good and poor outcomes, respectively. miR-375 could be used in all recipients irrespective of GAD65 autoantibody status (n = 46), with levels <1.4 pmol/L (LR = 0.14) or >7.6 pmol/L (LR = 9.53) as dual thresholds. The posttransplant surge of unmethylated insulin DNA was inconsistent and unrelated to outcome. Combined measurement of these three biomarkers was also tested as liquid biopsy for ß cell death during 2-month follow-up; incidental surges of GAD65, miR-375, and (un)methylated insulin DNA, alone or combined, were confidently detected but could not be related to outcome. Conclusions: GAD65 and miR-375 performed equally well in quantifying early graft destruction and predicting graft outcome, outperforming unmethylated insulin DNA.


Subject(s)
Diabetes Mellitus, Type 1/surgery , Glutamate Decarboxylase/blood , Graft Rejection/diagnosis , Insulin/blood , Islets of Langerhans Transplantation/adverse effects , MicroRNAs/blood , Adult , Biomarkers , DNA Methylation , Follow-Up Studies , Graft Rejection/blood , Humans , Insulin/genetics , Middle Aged , Postoperative Period , Prognosis
6.
Diabetes ; 67(12): 2640-2649, 2018 12.
Article in English | MEDLINE | ID: mdl-30305364

ABSTRACT

Alginate (Alg)-encapsulated porcine islet cell grafts are developed to overcome limitations of human islet transplantation. They can generate functional implants in animals when prepared from fetal, perinatal, and adult pancreases. Implants have not yet been examined for efficacy to establish sustained, metabolically adequate functional ß-cell mass (FBM) in comparison with human islet cells. This study in immune-compromised mice demonstrates that subcutaneous implants of Alg-encapsulated porcine prenatal islet cells with 4 × 105 ß-cells form, over 10 weeks, a FBM that results in glucose-induced plasma C-peptide >2 ng/mL and metabolic control over the following 10 weeks, with higher efficiency than nonencapsulated, while failing in peritoneum. This intracapsular FBM formation involves ß-cell replication, increasing number fourfold, and maturation toward human adult ß-cells. Subcutaneous Alg-encapsulated human islet cells with similar ß-cell number establish implants with plasma C-peptide >2 ng/mL for the first 10 weeks, with nonencapsulated cells failing; their ß-cells do not replicate but progressively die (>70%), explaining C-peptide decline and insufficient metabolic control. An Alg matrix thus helps establish ß-cell functions in subcutis. It allows formation of sustained metabolically adequate FBM by immature porcine ß-cells with proliferative activity but not by human adult islet cells. These findings define conditions for evaluating its immune-protecting properties.


Subject(s)
Alginates , C-Peptide/blood , Insulin-Secreting Cells/metabolism , Insulin/blood , Islets of Langerhans Transplantation/methods , Animals , Capsules , Humans , Mice , Swine
7.
Diabetologia ; 61(7): 1623-1632, 2018 07.
Article in English | MEDLINE | ID: mdl-29679103

ABSTRACT

AIMS/HYPOTHESIS: HLA-A*24 carriership hampers achievement of insulin independence in islet allograft recipients. However, less than half of those who fail to achieve insulin independence carry the allele. We investigated whether genetic polymorphism at the recipients' zinc transporter 8-encoding SLC30A8 gene (rs13266634) could complement their HLA-A*24 status in predicting functional graft outcome. METHODS: We retrospectively analysed data of a hospital-based patient cohort followed for 18 months post transplantation. Forty C-peptide-negative type 1 diabetic individuals who received >2 million beta cells (>4000 islet equivalents) per kg body weight in one or two intraportal implantations under similar immunosuppression were genotyped for SLC30A8. Outcome measurements included achievement and maintenance of graft function. Metabolic benefit was defined as <25% CV of fasting glycaemia in the presence of >331 pmol/l C-peptide, in addition to achievement of insulin independence and maintenance of C-peptide positivity. RESULTS: In multivariate analysis, HLA-A*24 positivity, presence of SLC30A8 CT or TT genotypes and BMI more than or equal to the group median (23.9 kg/m2) were independently associated with failure to achieve insulin independence (p = 0.015-0.046). The risk increased with the number of factors present (p < 0.001). High BMI interacted with SLC30A8 T allele carriership to independently predict difficulty in achieving graft function with metabolic benefit (p = 0.015). Maintenance of C-peptide positivity was mainly associated with older age at the time of implantation. Only HLA-A*24 carriership independently predicted failure to maintain acceptable graft function once achieved (p = 0.012). CONCLUSIONS/INTERPRETATION: HLA-A*24, the SLC30A8 T allele and high BMI are associated with poor graft outcome and should be considered in the interpretation of future transplantation trials. TRIAL REGISTRATION: ClinicalTrials.gov NCT00798785 and NCT00623610.


Subject(s)
Blood Glucose/metabolism , Body Mass Index , Diabetes Mellitus, Type 1/surgery , HLA-A24 Antigen/genetics , Insulin-Secreting Cells/transplantation , Islets of Langerhans Transplantation/adverse effects , Polymorphism, Genetic , Zinc Transporter 8/genetics , Allografts , Biomarkers/blood , Blood Glucose/drug effects , Clinical Trials as Topic , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Female , HLA-A24 Antigen/immunology , Humans , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Male , Middle Aged , Recovery of Function , Retrospective Studies , Risk Factors , Treatment Outcome
8.
Diabetes Care ; 41(5): 1076-1083, 2018 05.
Article in English | MEDLINE | ID: mdl-29545461

ABSTRACT

OBJECTIVE: We investigated the effect of HLA class I risk alleles on disease progression in various phases of subclinical islet autoimmunity in first-degree relatives of patients with type 1 diabetes. RESEARCH DESIGN AND METHODS: A registry-based group of siblings/offspring (aged 0-39 years) was monitored from single- to multiple-autoantibody positivity (n = 267) and from multiple-autoantibody positivity to clinical onset (n = 252) according to HLA-DQ, -A*24, -B*18, and -B*39 status. Genetic markers were determined by PCR sequence-specific oligotyping. RESULTS: Unlike HLA-B*18 or -B*39, HLA-A*24 was associated with delayed progression from single- to multiple-autoantibody positivity (P = 0.009) but not to type 1 diabetes. This occurred independently from older age (P < 0.001) and absence of HLA-DQ2/DQ8 or -DQ8 (P < 0.001 and P = 0.003, respectively), and only in the presence of GAD autoantibodies. In contrast, HLA-A*24 was associated with accelerated progression from multiple-autoantibody positivity to clinical onset (P = 0.006), but its effects were restricted to HLA-DQ8+ relatives with IA-2 or zinc transporter 8 autoantibodies (P = 0.002). HLA-B*18, but not -B*39, was also associated with more rapid progression, but only in HLA-DQ2 carriers with double positivity for GAD and insulin autoantibodies (P = 0.004). CONCLUSIONS: HLA-A*24 predisposes to a delayed antigen spreading of humoral autoimmunity, whereas HLA-A*24 and -B*18 are associated with accelerated progression of advanced subclinical autoimmunity in distinct risk groups. The relation of these alleles to the underlying disease process requires further investigation. Their typing should be relevant for the preparation and interpretation of observational and interventional studies in asymptomatic type 1 diabetes.


Subject(s)
Autoantibodies/blood , Autoimmunity/genetics , Diabetes Mellitus, Type 1/pathology , HLA-A24 Antigen/genetics , HLA-B18 Antigen/genetics , HLA-DQ Antigens/genetics , Islets of Langerhans/immunology , Adolescent , Adult , Child , Child, Preschool , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Disease Progression , Female , Genetic Predisposition to Disease , Genotype , Humans , Infant , Infant, Newborn , Insulin Antibodies/blood , Male , Registries , Risk Factors , Young Adult
9.
Stem Cell Reports ; 10(3): 739-750, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29503087

ABSTRACT

Human stem cells represent a potential source for implants that replace the depleted functional beta cell mass (FBM) in diabetes patients. Human embryonic stem cell-derived pancreatic endoderm (hES-PE) can generate implants with glucose-responsive beta cells capable of reducing hyperglycemia in mice. This study with device-encapsulated hES-PE (4 × 106 cells/mouse) determines the biologic characteristics at which implants establish metabolic control during a 50-week follow-up. A metabolically adequate FBM was achieved by (1) formation of a sufficient beta cell number (>0.3 × 106/mouse) at >50% endocrine purity and (2) their maturation to a functional state comparable with human pancreatic beta cells, as judged by their secretory responses during perifusion, their content in typical secretory vesicles, and their nuclear NKX6.1-PDX1-MAFA co-expression. Assessment of FBM in implants and its correlation with in vivo metabolic markers will guide clinical translation of stem cell-derived grafts in diabetes.


Subject(s)
Embryonic Stem Cells/metabolism , Embryonic Stem Cells/physiology , Endoderm/metabolism , Endoderm/physiology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/physiology , Animals , Cell Line , Glucose/metabolism , Homeodomain Proteins/metabolism , Humans , Maf Transcription Factors, Large/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Trans-Activators/metabolism
10.
Diabetes Care ; 40(8): 1065-1072, 2017 08.
Article in English | MEDLINE | ID: mdl-28701370

ABSTRACT

OBJECTIVE: We investigated whether islet autoantibody profile, HLA-DQ genotype, and age influenced a 20-year progression to diabetes from first autoantibody positivity (autoAb+) in first-degree relatives of patients with type 1 diabetes. RESEARCH DESIGN AND METHODS: Persistently islet autoAb+ siblings and offspring (n = 462) under 40 years of age were followed by the Belgian Diabetes Registry. AutoAbs against insulin (IAA), GAD (GADA), IA-2 antigen (IA-2A), and zinc transporter 8 (ZnT8A) were determined by radiobinding assay. RESULTS: The 20-year progression rate of multiple-autoAb+ relatives (n = 194) was higher than that for single-autoAb+ participants (n = 268) (88% vs. 54%; P < 0.001). Relatives positive for IAA and GADA (n = 54) progressed more slowly than double-autoAb+ individuals carrying IA-2A and/or ZnT8A (n = 38; P = 0.001). In multiple-autoAb+ relatives, Cox regression analysis identified the presence of IA-2A or ZnT8A as the only independent predictors of more rapid progression to diabetes (P < 0.001); in single-autoAb+ relatives, it identified younger age (P < 0.001), HLA-DQ2/DQ8 genotype (P < 0.001), and IAA (P = 0.028) as independent predictors of seroconversion to multiple positivity for autoAbs. In time-dependent Cox regression, younger age (P = 0.042), HLA-DQ2/DQ8 genotype (P = 0.009), and the development of additional autoAbs (P = 0.012) were associated with more rapid progression to diabetes. CONCLUSIONS: In single-autoAb+ relatives, the time to multiple-autoAb positivity increases with age and the absence of IAA and HLA-DQ2/DQ8 genotype. The majority of multiple-autoAb+ individuals progress to diabetes within 20 years; this occurs more rapidly in the presence of IA-2A or ZnT8A, regardless of age, HLA-DQ genotype, and number of autoAbs. These data may help to refine the risk stratification of presymptomatic type 1 diabetes.


Subject(s)
Autoantibodies/blood , Diabetes Mellitus, Type 1/blood , Disease Progression , HLA-DQ Antigens/genetics , Registries , Adolescent , Adult , Belgium , Child , Child, Preschool , Diabetes Mellitus, Type 1/genetics , Female , Follow-Up Studies , Humans , Infant , Insulin/blood , Male , Proportional Hazards Models , Risk Factors , Surveys and Questionnaires , Young Adult , Zinc Transporter 8/blood
11.
Diabetes Care ; 39(6): 1060-4, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27208324

ABSTRACT

OBJECTIVE: We investigated whether changes in islet autoantibody profile and presence of HLA risk markers, reported to predict rapid ß-cell loss in pre-type 1 diabetes, associate with poor functional outcome in islet allograft recipients. RESEARCH DESIGN AND METHODS: Forty-one patients received ≥2.3 million ß-cells/kg body wt in one to two intraportal implantations. Outcome after 6-18 months was assessed by C-peptide (random and stimulated), insulin dose, and HbA1c. RESULTS: Patients carrying HLA-A*24-positive or experiencing a significant autoantibody surge within 6 months after the first transplantation (n = 19) had lower C-peptide levels (P ≤ 0.003) and higher insulin needs (P < 0.001) despite higher HbA1c levels (P ≤ 0.018). They became less often insulin independent (16% vs. 68%, P = 0.002) and remained less often C-peptide positive (47% vs. 100%, P < 0.001) than recipients lacking both risk factors. HLA-A*24 positivity or an autoantibody surge predicted insulin dependence (P = 0.007). CONCLUSIONS: HLA-A*24 and early autoantibody surge after islet implantation associate with poor functional graft outcome.


Subject(s)
Autoantibodies/immunology , Diabetes Mellitus, Type 1/therapy , HLA-A24 Antigen/genetics , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Islets of Langerhans Transplantation , Adult , Allografts , C-Peptide/metabolism , Cation Transport Proteins/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Female , Glutamate Decarboxylase/immunology , Humans , Insulin-Secreting Cells , Male , Middle Aged , Transplantation, Homologous , Treatment Outcome , Zinc Transporter 8
12.
Diabetologia ; 58(12): 2753-64, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26409458

ABSTRACT

AIMS/HYPOTHESIS: We examined whether measures of glycaemic variability (GV), assessed by continuous glucose monitoring (CGM) and self-monitoring of blood glucose (SMBG), can complement or replace measures of beta cell function and insulin action in detecting the progression of preclinical disease to type 1 diabetes. METHODS: Twenty-two autoantibody-positive (autoAb(+)) first-degree relatives (FDRs) of patients with type 1 diabetes who were themselves at high 5-year risk (50%) for type 1 diabetes underwent CGM, a hyperglycaemic clamp test and OGTT, and were followed for up to 31 months. Clamp variables were used to estimate beta cell function (first-phase [AUC5-10 min] and second-phase [AUC120-150 min] C-peptide release) combined with insulin resistance (glucose disposal rate; M 120-150 min). Age-matched healthy volunteers (n = 20) and individuals with recent-onset type 1 diabetes (n = 9) served as control groups. RESULTS: In autoAb(+) FDRs, M 120-150 min below the 10th percentile (P10) of controls achieved 86% diagnostic efficiency in discriminating between normoglycaemic FDRs and individuals with (impending) dysglycaemia. M 120-150 min outperformed AUC5-10 min and AUC120-150 min C-peptide below P10 of controls, which were only 59-68% effective. Among GV variables, CGM above the reference range was better at detecting (impending) dysglycaemia than elevated SMBG (77-82% vs 73% efficiency). Combined CGM measures were equally efficient as M 120-150 min (86%). Daytime GV variables were inversely correlated with clamp variables, and more strongly with M 120-150 min than with AUC5-10 min or AUC120-150 min C-peptide. CONCLUSIONS/INTERPRETATION: CGM-derived GV and the glucose disposal rate, reflecting both insulin secretion and action, outperformed SMBG and first- or second-phase AUC C-peptide in identifying FDRs with (impending) dysglycaemia or diabetes. Our results indicate the feasibility of developing minimally invasive CGM-based criteria for close metabolic monitoring and as outcome measures in trials.


Subject(s)
Blood Glucose/analysis , Diabetes Mellitus, Type 1/blood , Glucose Clamp Technique , Hyperglycemia/blood , Adolescent , Adult , Area Under Curve , Blood Glucose Self-Monitoring , C-Peptide/blood , Child , Female , Glucose Tolerance Test , Glycated Hemoglobin/analysis , Healthy Volunteers , Humans , Insulin-Secreting Cells/metabolism , Male , Young Adult
13.
Diabetes Care ; 38(4): 644-51, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25583753

ABSTRACT

OBJECTIVE: Immune intervention trials in recent-onset type 1 diabetes would benefit from biomarkers associated with good therapeutic response. In the previously reported randomized placebo-controlled anti-CD3 study (otelixizumab; GlaxoSmithKline), we tested the hypothesis that specific diabetes autoantibodies might serve this purpose. RESEARCH DESIGN AND METHODS: In the included patients (n = 40 otelixizumab, n = 40 placebo), ß-cell function was assessed as area under the curve (AUC) C-peptide release during a hyperglycemic glucose clamp at baseline (median duration of insulin treatment: 6 days) and every 6 months until 18 months after randomization. (Auto)antibodies against insulin (I[A]A), GAD (GADA), IA-2 (IA-2A), and ZnT8 (ZnT8A) were determined on stored sera by liquid-phase radiobinding assay. RESULTS: At baseline, only better preserved AUC C-peptide release and higher levels of IAA were associated with better preservation of ß-cell function and lower insulin needs under anti-CD3 treatment. In multivariate analysis, IAA (P = 0.022) or the interaction of IAA and C-peptide (P = 0.013) independently predicted outcome together with treatment. During follow-up, good responders to anti-CD3 treatment (i.e., IAA(+) participants with relatively preserved ß-cell function [≥ 25% of healthy control subjects]) experienced a less pronounced insulin-induced rise in I(A)A and lower insulin needs. GADA, IA-2A, and ZnT8A levels were not influenced by anti-CD3 treatment, and their changes showed no relation to functional outcome. CONCLUSIONS: There is important specificity of IAA among other diabetes autoantibodies to predict good therapeutic response of recent-onset type 1 diabetic patients to anti-CD3 treatment. If confirmed, future immune intervention trials in type 1 diabetes should consider both relatively preserved functional ß-cell mass and presence of IAA as inclusion criteria.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/drug therapy , Insulin Antibodies/blood , Insulin-Secreting Cells/drug effects , Adolescent , Adult , Biomarkers/blood , Child , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/physiopathology , Female , Humans , Insulin/therapeutic use , Insulin-Secreting Cells/physiology , Male , Prognosis , Time Factors , Treatment Outcome , Young Adult
14.
J Clin Endocrinol Metab ; 100(2): 551-60, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25405499

ABSTRACT

CONTEXT AND OBJECTIVE: In preparation of future prevention trials, we aimed to identify predictors of 3-year diabetes onset among oral glucose tolerance test (OGTT)- and hyperglycemic clamp-derived metabolic markers in persistently islet autoantibody positive (autoAb(+)) offspring and siblings of patients with type 1 diabetes (T1D). DESIGN: The design is a registry-based study. SETTING: Functional tests were performed in a hospital setting. PARTICIPANTS: Persistently autoAb(+) first-degree relatives of patients with T1D (n = 81; age 5-39 years). MAIN OUTCOME MEASURES: We assessed 3-year predictive ability of OGTT- and clamp-derived markers using receiver operating characteristics (ROC) and Cox regression analysis. Area under the curve of clamp-derived first-phase C-peptide release (AUC(5-10 min); min 5-10) was determined in all relatives and second-phase release (AUC(120-150 min); min 120-150) in those aged 12-39 years (n = 62). RESULTS: Overall, the predictive ability of AUC(5-10 min) was better than that of peak C-peptide, the best predictor among OGTT-derived parameters (ROC-AUC [95%CI]: 0.89 [0.80-0.98] vs 0.81 [0.70-0.93]). Fasting blood glucose (FBG) and AUC(5-10 min) provided the best combination of markers for prediction of diabetes within 3 years; (ROC-AUC [95%CI]: 0.92 [0.84-1.00]). In multivariate Cox regression analysis, AUC(5-10 min)) (P = .001) was the strongest independent predictor and interacted significantly with all tested OGTT-derived parameters. AUC(5-10 min) below percentile 10 of controls was associated with 50-70% progression to T1D regardless of age. Similar results were obtained for AUC(120-150 min). CONCLUSIONS: Clamp-derived first-phase C-peptide release can be used as an efficient and simple screening strategy in persistently autoAb(+) offspring and siblings of T1D patients to predict impending diabetes.


Subject(s)
Autoantibodies/blood , Child of Impaired Parents , Diabetes Mellitus, Type 1/blood , Siblings , Adolescent , Adult , Child , Child, Preschool , Diabetes Mellitus, Type 1/immunology , Female , Glucose Intolerance , Glucose Tolerance Test , Humans , Insulin/blood , Male , Predictive Value of Tests , ROC Curve , Young Adult
15.
Diabetes Res Clin Pract ; 103(1): 97-105, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24332797

ABSTRACT

AIMS: We investigated the prevalence of diabetes autoantibodies (Abs) in Cameroonian patients and controls, assessed their contribution in disease classification and compared results with data from Belgium. METHODS: Abs against GAD (GADA), IA-2 (IA-2A) and zinc transporter 8 (ZnT8A) were assessed in 302 recently diagnosed Cameroonian patients with diabetes and 184 control subjects without diabetes aged below 40 years. RESULTS: Only 27 (9%) Cameroonian patients were younger than 15 years. Overall, 29% of patients presented at least one diabetes-associated antibody vs 9% in healthy controls (24% vs 7% for GADA (p<0.001), 10% vs 3% for IA-2A (p<0.006), 4% vs 2% for ZnT8A). Ab(+) patients had lower C-peptide levels (p<0.001), were more often insulin-treated (p<0.002) and were as frequently diagnosed with type 1 diabetes as Ab(-) patients. Only 43% of Ab(+) patients aged 15-39 years were clinically classified as having type 1 diabetes in Cameroon vs 96% in Belgium (p<0.001). Not one Ab(+) Cameroonian patient carried HLA-DQ2/DQ8 genotype vs 23% of Belgian Ab(+) patients (p<0.001). Younger age at diagnosis and antibody positivity were independent predictors of insulin therapy. Ab(+) Cameroonian patients were older (p<0.001), had higher BMI (p<0.001) and lower Ab titers than Belgian Ab(+) patients. In ketonuric patients, prevalence of autoantibodies was similar as in non-ketonuric patients. CONCLUSIONS: In Cameroonian patients with diabetes aged under 40 years, antibody-positivity is not clearly related to disease phenotype, but may help predict the need for insulin treatment.


Subject(s)
Autoantibodies/blood , Biomarkers/blood , Cation Transport Proteins/immunology , Diabetes Mellitus, Type 1/immunology , Glutamate Decarboxylase/immunology , Receptor-Like Protein Tyrosine Phosphatases, Class 8/immunology , Adolescent , Adult , Belgium/epidemiology , Cameroon/epidemiology , Case-Control Studies , Child , Child, Preschool , Diabetes Mellitus, Type 1/epidemiology , Female , Humans , Infant , Infant, Newborn , Male , Prevalence , Young Adult , Zinc Transporter 8
16.
Expert Rev Clin Immunol ; 9(12): 1173-83, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24215407

ABSTRACT

Type 1 diabetes is an incurable disease associated with risk of serious acute and chronic complications. It is caused by a marked loss in insulin-producing beta cells. Immune intervention at clinical onset can transiently suppress a further decline in residual functional beta cell mass, particularly in young patients with a higher residual insulin producing capacity at start of treatment. It might achieve a higher effect during the preclinical phase when the beta cell mass is not yet severely affected. Such prevention trials can be prepared by identifying individuals at very high risk to develop diabetes within 3 years and presenting signs of declining, yet relatively preserved, functional beta cell mass. This article reviews biomarker screening strategies to select these participants and discusses developments that can facilitate rapid and straightforward conclusions from novel clinical studies.


Subject(s)
Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/immunology , Immunotherapy/trends , Insulin-Secreting Cells/drug effects , Animals , Biomarkers/metabolism , Clinical Trials as Topic , Diabetes Mellitus, Type 1/prevention & control , Disease Progression , Humans , Insulin-Secreting Cells/pathology , Patient Selection , Predictive Value of Tests , Prognosis , Risk
17.
Transplantation ; 96(12): 1026-33, 2013 Dec 27.
Article in English | MEDLINE | ID: mdl-24056625

ABSTRACT

BACKGROUND: Rapid revascularization of islet cell implants is important for engraftment and subsequent survival and function. Development of an adequate vascular network is expected to allow adaptive growth of the ß-cell mass. The present study compares omentum and kidney capsule as sites for growth and differentiation of immature ß-cell grafts. METHODS: Perinatal porcine islet cell grafts were implanted in omentum or under kidney capsule of nondiabetic nude rats. Implants were compared over 10 weeks for their respective growth, cellular composition, number and size of ß cells, their proliferative activity, and implant blood vessel density. RESULTS: In both sites, the ß-cell volume increased fourfold between weeks 1 and 10 reflecting a rise in ß-cell number. In the omental implants, however, the cellular insulin reserves and the percent of proliferating cells were twofold higher than in kidney implants. In parallel, the blood vessel density in omental implants increased twofold, reaching a density comparable with islets in adult pig pancreas. A positive correlation was found between the percent bromodeoxyuridine-positive ß cells and the vessel density. CONCLUSIONS: Growth of the ß-cell volume proceeds similarly in the omentum and under the kidney capsule. However, the omentum leads to higher insulin reserves and an increased pool of proliferating cells, which might be related to a more extended vascular network. Our observations support the omentum as an alternative site for immature porcine islet cells, with beneficial effects on proliferation and implant revascularization.


Subject(s)
Insulin-Secreting Cells/cytology , Insulin/metabolism , Islets of Langerhans Transplantation/methods , Kidney/pathology , Omentum/pathology , Animals , Blood Vessels/pathology , Bromodeoxyuridine , Cell Proliferation , Cell Survival , Glucagon-Secreting Cells/cytology , Immunohistochemistry , Male , Rats , Rats, Nude , Rats, Wistar , Swine
18.
Endocrinology ; 154(5): 1934-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23525244

ABSTRACT

The limitations of current rat C-peptide assays led us to develop a time-resolved fluorescence immunoassay for measurements in plasma, incubation media, and tissue/cell extracts. The assay uses 2 monoclonal antibodies, binding to different parts of the C-peptide molecule, and allowing, respectively, capture of the peptide and its detection by europium-labeled streptavidin. It is performed on 25-µL samples for a dynamic range from 66pM up to 3900pM C-peptide and displays over 95% recovery of added peptide in the range of 111pM to 2786pM. Its inter- and intra-assay coefficients of variations are, respectively, lower than 7.6% and 4.8%. Cross-reactivities by rat insulin and by human and porcine C-peptide are negligible, and cross-reactivity by mouse C-peptide is 6% ± 2%. The assay has been validated for in vivo and in vitro measurements of C-peptide release and cellular content. Release patterns were similar to those for insulin and occurred in equimolar concentrations for both peptides. The molar C-peptide contents in purified ß-cells and isolated islets were similar to the corresponding insulin contents. This was also the case for pancreatic extracts containing protease inhibitors.


Subject(s)
C-Peptide/analysis , Fluoroimmunoassay/methods , Insulin-Secreting Cells/metabolism , Animals , C-Peptide/immunology , C-Peptide/metabolism , Cells, Cultured , Fluorescence , Humans , Insulin-Secreting Cells/chemistry , Mice , Rats , Sensitivity and Specificity , Species Specificity , Swine
19.
PLoS One ; 6(9): e24134, 2011.
Article in English | MEDLINE | ID: mdl-21912665

ABSTRACT

BACKGROUND AND METHODOLOGY: The aim of this study was to establish a gene expression blueprint of pancreatic beta cells conserved from rodents to humans and to evaluate its applicability to assess shifts in the beta cell differentiated state. Genome-wide mRNA expression profiles of isolated beta cells were compared to those of a large panel of other tissue and cell types, and transcripts with beta cell-abundant and -selective expression were identified. Iteration of this analysis in mouse, rat and human tissues generated a panel of conserved beta cell biomarkers. This panel was then used to compare isolated versus laser capture microdissected beta cells, monitor adaptations of the beta cell phenotype to fasting, and retrieve possible conserved transcriptional regulators. PRINCIPAL FINDINGS: A panel of 332 conserved beta cell biomarker genes was found to discriminate both isolated and laser capture microdissected beta cells from all other examined cell types. Of all conserved beta cell-markers, 15% were strongly beta cell-selective and functionally associated to hormone processing, 15% were shared with neuronal cells and associated to regulated synaptic vesicle transport and 30% with immune plus gut mucosal tissues reflecting active protein synthesis. Fasting specifically down-regulated the latter cluster, but preserved the neuronal and strongly beta cell-selective traits, indicating preserved differentiated state. Analysis of consensus binding site enrichment indicated major roles of CREB/ATF and various nutrient- or redox-regulated transcription factors in maintenance of differentiated beta cell phenotype. CONCLUSIONS: Conserved beta cell marker genes contain major gene clusters defined by their beta cell selectivity or by their additional abundance in either neural cells or in immune plus gut mucosal cells. This panel can be used as a template to identify changes in the differentiated state of beta cells.


Subject(s)
Conserved Sequence , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Multigene Family/genetics , Oligonucleotide Array Sequence Analysis , Transcriptome/genetics , Animals , Cell Differentiation/genetics , Fasting/metabolism , Genetic Markers/genetics , Humans , Male , Mice , Neurons/cytology , Neurons/metabolism , Phenotype , Rats , Real-Time Polymerase Chain Reaction , Transcription Factors/metabolism
20.
Diabetes Care ; 34(8): 1760-5, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21715527

ABSTRACT

OBJECTIVE: We investigated whether measuring autoantibodies against zinc transporter 8 (ZnT8A) and IA-2ß (IA-2ßA) may improve classification of new-onset type 1 diabetic patients based on detection of autoantibodies against insulin (IAA), GAD (GADA), and IA-2 (IA-2A). In addition, we studied the correlation of IA-2ßA and ZnT8A with other biological and demographic variables. RESEARCH DESIGN AND METHODS: Circulating autoantibodies were determined by liquid-phase radiobinding assays from 761 healthy control subjects and 655 new-onset (<1 week insulin) diabetic patients (aged 0-39 years) with clinical type 1 diabetes phenotype consecutively recruited by the Belgian Diabetes Registry. RESULTS: At diagnosis, IA-2ßA and ZnT8A prevalences were 41 and 58%, respectively. In IAA-negative, GADA-negative, and IA-2A-negative patients, one IA-2ßA-positive and eleven ZnT8A-positive individuals were identified at the expense of eight and seven additional positive control subjects (1%), respectively, for each test. ZnT8A or IA-2ßA screening increased (P < 0.001; McNemar) the number of patients with ≥2 antibodies both under (from 78 to 87% for ZnT8A and 82% for IA-2ßA) and above age 15 (from 51 to 63% for ZnT8A and 56% for IA-2ßA) versus 0% in control subjects. IA-2ßA and ZnT8A were preferentially associated with IA-2A, and with younger age at diagnosis. Unlike ZnT8A, IA-2ßA levels were positively correlated with HLA-DQ8 and negatively with HLA-DQ2. ZnT8A could replace IAA for classification of patients above age 10 without loss of sensitivity or specificity. CONCLUSIONS: ZnT8A, and to a lesser degree IA-2ßA, may usefully complement GADA, IA-2A, and IAA for classifying insulin-treated diabetes under age 40 years.


Subject(s)
Autoantibodies/immunology , Cation Transport Proteins/immunology , Diabetes Mellitus, Type 1/classification , Diabetes Mellitus, Type 1/immunology , Receptor-Like Protein Tyrosine Phosphatases, Class 8/immunology , Adolescent , Adult , Child , Diabetes Mellitus, Type 1/diagnosis , Female , Humans , Male , Young Adult , Zinc Transporter 8
SELECTION OF CITATIONS
SEARCH DETAIL
...