Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 152
Filter
Add more filters

Publication year range
1.
PLoS Comput Biol ; 19(2): e1010914, 2023 02.
Article in English | MEDLINE | ID: mdl-36848379

ABSTRACT

Environmentally induced or epigenetic-related beta-cell dysfunction and insulin resistance play a critical role in the progression to diabetes. We developed a mathematical modeling framework capable of studying the progression to diabetes incorporating various diabetogenic factors. Considering the heightened risk of beta-cell defects induced by obesity, we focused on the obesity-diabetes model to further investigate the influence of obesity on beta-cell function and glucose regulation. The model characterizes individualized glucose and insulin dynamics over the span of a lifetime. We then fit the model to the longitudinal data of the Pima Indian population, which captures both the fluctuations and long-term trends of glucose levels. As predicted, controlling or eradicating the obesity-related factor can alleviate, postpone, or even reverse diabetes. Furthermore, our results reveal that distinct abnormalities of beta-cell function and levels of insulin resistance among individuals contribute to different risks of diabetes. This study may encourage precise interventions to prevent diabetes and facilitate individualized patient treatment.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Humans , Diabetes Mellitus, Type 2/complications , Insulin Resistance/physiology , Insulin , Obesity/complications , Glucose , Blood Glucose
2.
Diabetes Obes Metab ; 26(2): 463-472, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37867175

ABSTRACT

AIM: This study compared the 5-year incidence rate of macrovascular and microvascular complications for tirzepatide, semaglutide and insulin glargine in individuals with type 2 diabetes, using the Building, Relating, Assessing, and Validating Outcomes (BRAVO) diabetes simulation model. RESEARCH DESIGN AND METHODS: This study was a 5-year SURPASS-2 trial extrapolation, with an insulin glargine arm added as an additional comparator. The 1-year treatment effects of tirzepatide (5, 10 or 15 mg), semaglutide (1 mg) and insulin glargine on glycated haemoglobin, systolic blood pressure, low-density lipoprotein and body weights were obtained from the SUSTAIN-4 and SURPASS-2 trials. We used the BRAVO model to predict 5-year complications for each study arm under two scenarios: the 1-year treatment effects persisted (optimistic) or diminished to none in 5 years (conservative). RESULTS: When compared with insulin glargine, we projected a 5-year risk reduction in cardiovascular adverse events [rate ratio (RR) 0.64, 95% confidence interval (CI) 0.61-0.67] and microvascular composite (RR 0.67, 95% CI 0.64-0.70) with 15 mg tirzepatide, and 5-year risk reduction in cardiovascular adverse events (RR 0.75, 95% CI 0.72-0.79) and microvascular composite (RR 0.79, 95% CI 0.76-0.82) with semaglutide (1 mg) under an optimistic scenario. Lower doses of tirzepatide also had similar, albeit smaller benefits. Treatment effects for tirzepatide and semaglutide were smaller but still significantly higher than insulin glargine under a conservative scenario. The 5-year risk reduction in diabetes-related complication events and mortality for the 15 mg tirzepatide compared with insulin glargine ranged from 49% to 10% under an optimistic scenario, which was reduced by 17%-33% when a conservative scenario was assumed. CONCLUSION: With the use of the BRAVO diabetes model, tirzepatide and semaglutide exhibited potential to reduce the risk of macrovascular and microvascular complications among individuals with type 2 diabetes, compared with insulin glargine in a 5-year window. Based on the current modelling assumptions, tirzepatide (15 mg) may potentially outperform semaglutide (1 mg). While the BRAVO model offered insights, the long-term cardiovascular benefit of tirzepatide should be further validated in a prospective clinical trial.


Subject(s)
Diabetes Complications , Diabetes Mellitus, Type 2 , Humans , Diabetes Complications/drug therapy , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/adverse effects , Insulin Glargine/adverse effects , Prospective Studies
3.
Am J Pathol ; 191(3): 454-462, 2021 03.
Article in English | MEDLINE | ID: mdl-33307036

ABSTRACT

Emerging data suggest that type 1 diabetes affects not only the ß-cell-containing islets of Langerhans, but also the surrounding exocrine compartment. Using digital pathology, machine learning algorithms were applied to high-resolution, whole-slide images of human pancreata to determine whether the tissue composition in individuals with or at risk for type 1 diabetes differs from those without diabetes. Transplant-grade pancreata from organ donors were evaluated from 16 nondiabetic autoantibody-negative controls, 8 nondiabetic autoantibody-positive subjects with increased type 1 diabetes risk, and 19 persons with type 1 diabetes (0 to 12 years' duration). HALO image analysis algorithms were implemented to compare architecture of the main pancreatic duct as well as cell size, density, and area of acinar, endocrine, ductal, and other nonendocrine, nonexocrine tissues. Type 1 diabetes was found to affect exocrine area, acinar cell density, and size, whereas the type of difference correlated with the presence or absence of insulin-positive cells remaining in the pancreas. These changes were not observed before disease onset, as indicated by modeling cross-sectional data from pancreata of autoantibody-positive subjects and those diagnosed with type 1 diabetes. These data provide novel insights into anatomic differences in type 1 diabetes pancreata and demonstrate that machine learning can be adapted for the evaluation of disease processes from cross-sectional data sets.


Subject(s)
Algorithms , Autoantibodies/immunology , Diabetes Mellitus, Type 1/pathology , Image Processing, Computer-Assisted/methods , Machine Learning , Pancreas/pathology , Adolescent , Autoantibodies/blood , Case-Control Studies , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Female , Humans , Insulin/analysis , Pancreas/immunology , Pancreas/metabolism , Tissue Donors
4.
Diabetologia ; 64(4): 836-844, 2021 04.
Article in English | MEDLINE | ID: mdl-33496819

ABSTRACT

AIMS/HYPOTHESIS: We aimed to compare characteristics of individuals identified in the peri-diagnostic range by Index60 (composite glucose and C-peptide measure) ≥2.00, 2 h OGTT glucose ≥11.1 mmol/l, or both. METHODS: We studied autoantibody-positive participants in the Type 1 Diabetes TrialNet Pathway to Prevention study who, at their baseline OGTT, had 2 h blood glucose ≥11.1 mmol/l and/or Index60 ≥2.00 (n = 354, median age = 11.2 years, age range = 1.7-46.6; 49% male, 83% non-Hispanic White). Type 1 diabetes-relevant characteristics (e.g., age, C-peptide, autoantibodies, BMI) were compared among three mutually exclusive groups: 2 h glucose ≥11.1 mmol/l and Index60 <2.00 [Glu(+), n = 76], 2 h glucose <11.1 mmol/l and Index60 ≥2.00 [Ind(+), n = 113], or both 2 h glucose ≥11.1 mmol/l and Index60 ≥2.00 [Glu(+)/Ind(+), n = 165]. RESULTS: Participants in Glu(+), vs those in Ind(+) or Glu(+)/Ind(+), were older (mean ages = 22.9, 11.8 and 14.7 years, respectively), had higher early (30-0 min) C-peptide response (1.0, 0.50 and 0.43 nmol/l), higher AUC C-peptide (2.33, 1.13 and 1.10 nmol/l), higher percentage of overweight/obesity (58%, 16% and 30%) (all comparisons, p < 0.0001), and a lower percentage of multiple autoantibody positivity (72%, 92% and 93%) (p < 0.001). OGTT-stimulated C-peptide and glucose patterns of Glu(+) differed appreciably from Ind(+) and Glu(+)/Ind(+). Progression to diabetes occurred in 61% (46/76) of Glu(+) and 63% (71/113) of Ind(+). Even though Index60 ≥2.00 was not a Pathway to Prevention diagnostic criterion, Ind(+) had a 4 year cumulative diabetes incidence of 95% (95% CI 86%, 98%). CONCLUSIONS/INTERPRETATION: Participants in the Ind(+) group had more typical characteristics of type 1 diabetes than participants in the Glu(+) did and were as likely to be diagnosed. However, unlike Glu(+) participants, Ind(+) participants were not identified at the baseline OGTT.


Subject(s)
Blood Glucose/metabolism , C-Peptide/blood , Decision Support Techniques , Diabetes Mellitus, Type 1/diagnosis , Glucose Tolerance Test , Islets of Langerhans/metabolism , Adolescent , Adult , Autoantibodies/blood , Biomarkers/blood , Child , Child, Preschool , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/immunology , Disease Progression , Early Diagnosis , Female , Humans , Infant , Islets of Langerhans/immunology , Male , Middle Aged , Predictive Value of Tests , Prognosis , Prospective Studies , Young Adult
5.
J Theor Biol ; 528: 110853, 2021 11 07.
Article in English | MEDLINE | ID: mdl-34358537

ABSTRACT

Current clinician practice for thyroid hormone regulation of patients is based upon guesswork and experience rather than quantified analysis, which exposes patients under longer risk and discomfort. To quantitatively analyze the thyroid regulation for patients of different thyroid states, we develop a two-dimensional mathematical model that can be applied to analyze the dynamic behaviors of thyroid hormones with or without drug intervention. The unified model can be employed to study the regulation of TSH (thyroid-stimulating hormone) and FT4 (free thyroxine) for euthyroid (normal thyroid) subjects, Hashimoto's thyroiditis, and Graves' disease patients, respectively. The results suggest that the level of TPOAb (thyroid peroxidase antibody) may be a factor determining whether the patient would progress from euthyroid state to subclinical or clinical hypothyroidism, and that increased TRAb (TSH receptor antibody) may lead Graves' disease to deteriorate from the early stage to overt hyperthyroidism. Given the early blood-test data, we demonstrate the feasibility for healthcare professionals to apply our model in choosing an appropriate dosage regimen for patients to achieve the desired TSH and FT4 levels within a specified time frame. This proposed model has the potential to optimize personalized treatment and shorten the therapeutic time for patients suffering from Hashimoto's thyroiditis and Graves' disease.


Subject(s)
Graves Disease , Precision Medicine , Autoantibodies , Graves Disease/drug therapy , Humans , Models, Theoretical , Thyroid Hormones
6.
Pediatr Diabetes ; 21(1): 120-127, 2020 02.
Article in English | MEDLINE | ID: mdl-31617648

ABSTRACT

Pronounced health disparities exist in type 1 diabetes (T1D) based on socioeconomic status (SES) yet there are a lack of programs designed to promote health equity for vulnerable communities. The All for ONE (Outreach, Networks, and Education) mentoring program was piloted pairing college students and publicly insured teenagers with T1D to assess feasibility as a possible intervention. There were 22 mentors recruited (mean age 20 ± 2 years; 17 [77%] females; mean HbA1c 8.4 ± 1.5%) and matched with mentees based on gender. There were 42 teens randomized to treatment and control groups including 22 teens in the treatment group (age 14 ± 2 years; 17 [77%] females; HbA1c 9.8 ± 2.3%) and 20 teens in the control group (age 14 ± 2 years; 15 [75%] females; HbA1c 8.9 ± 2.0%) followed over 9 months. Outcome measures included HbA1c and the Children's Hope Scale. The intervention included automated text reminders for blood glucose monitoring, text exchanges, social events with education, and clinic visits with mentors/mentees. Mean change in HbA1c for teens was +0.09% in the intervention group, compared with +0.28% in the control group (P = .61); college students had a reduction in HbA1c of -0.22% (P = .38). Treatment group teens had marked improvement in their hope for the future compared to control group teens (P = .04) and were more likely to attend clinic visits (P = .02). This program established feasibility for a model that could be replicated and modified for other types of settings. Additional research is warranted to study the potential long-term benefits of participating in the All for ONE mentoring program.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Health Promotion/methods , Health Status Disparities , Mentors , Adolescent , Age Factors , Blood Glucose Self-Monitoring , Child , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/psychology , Feasibility Studies , Female , Glycated Hemoglobin/metabolism , Hope , Humans , Male , Peer Group , Socioeconomic Factors , Young Adult
7.
Endocr Pract ; 26(12): 1505-1513, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33471743

ABSTRACT

OBJECTIVE: Type 1 diabetes (T1D) is characterized by autoimmune ß-cell destruction, but exocrine pancreas abnormalities may also play a role in the disease pathophysiology. Herein, we review the current evidence of exocrine damage in T1D and discuss its underlying pathophysiology, clinical evaluation, and treatment. METHOD: Extensive literature search was performed for "type 1 diabetes" and "exocrine dysfunction" on PubMed and Google Scholar databases. RESULTS: T1D pancreata are significantly smaller than controls, both in weight and volume. T cells, dendritic cells, neutrophils, and products of complement activation are seen in T1D exocrine tissues. Exocrine pancreas fibrosis, arteriosclerosis, fatty infiltration, and acinar atrophy are also observed on histology. Pancreatic exocrine insufficiency (PEI) can be assessed through direct exocrine testing, fecal elastase concentration, and measurement of serum exocrine enzymes. The prevalence of PEI in T1D varies by modality and study but is consistently greater than controls. The clinical relevance of PEI in T1D is debatable, as many patients with laboratory evidence of PEI are asymptomatic. However, in PEI-symptomatic patients reported benefits of pancreatic enzyme replacement therapy (PERT) include relief of gastrointestinal symptoms, improved quality of life, better glycemic control, and optimal nutrition. CONCLUSION: Exocrine pancreas abnormalities often occur in T1D. Whether exocrine dysfunction occurs simultaneously with ß-cell destruction, as a result of ß-cell loss, or as a combination of both remains to be definitively answered. In T1D with gastrointestinal complaints, PEI should be evaluated, usually via fecal elastase measurements. PERT is recommended for T1D patients with symptoms and laboratory evidence of PEI. ABBREVIATIONS: AAb+ = autoantibody positive; AAb- = autoantibody negative; FEC = fecal elastase concentration; PEI = pancreatic exocrine insufficiency; PERT = pancreatic enzyme replacement therapy; PP = pancreatic polypep-tide; T1D = type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1 , Exocrine Pancreatic Insufficiency , Pancreas, Exocrine , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/epidemiology , Enzyme Replacement Therapy , Exocrine Pancreatic Insufficiency/drug therapy , Exocrine Pancreatic Insufficiency/epidemiology , Exocrine Pancreatic Insufficiency/etiology , Humans , Pancreas , Quality of Life
8.
J Pediatr Psychol ; 44(1): 21-31, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30184209

ABSTRACT

Objectives: Type 1 diabetes (T1D) poses unique challenges to adherence-related behavior because of complex treatment regimens that vary by use of specific technologies. This study used objective data to determine (1) prevalence rates of adherence behaviors in adolescents with T1D, and (2) relationships between adherence and glycemic control. Methods: Data were downloaded for the past 30 consecutive days from glucose meters and multiple insulin pump models for 80 youth (11-17 years old; n = 40 on multiple daily injections (MDIs) and n = 40 on continuous subcutaneous insulin infusion [CSII]). Frequency of self-monitoring of blood glucose (SMBG; MDI and CSII users); carbohydrate entry (CSII users); daily insulin bolus delivery (CSII users); episodes of high, very high, and dangerously high hyperglycemia; and correction bolusing for hyperglycemia (CSII users) were calculated. Results: Participants completed SMBG ≥4 times/day on 46.13% of days (MDI users), 48.74% of days (CSII users nonmanual entries only), and 59.07% of days (CSII users; manual plus nonmanual entries). CSII users entered carbohydrates ≥3 times/day on 61.47% of days and bloused insulin ≥3 times/ day on 87.34% of days. Hyperglycemic readings were followed by a correction bolus in <70% of cases. Greater SMBG, carbohydrate entry, bolus insulin delivery, and correction bolusing for high and very high hyperglycemia predicted lower glycated hemoglobin (sample M = 8.74%, SD = 1.75%). Conclusions: Objective data from diabetes technology are helpful to differentiate adherence to specific domains of treatment but are complex in nature. Findings support a need for further research to elucidate predictive factors of suboptimal adherence in adolescents with T1D.


Subject(s)
Blood Glucose , Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/therapeutic use , Insulin Infusion Systems , Insulin/therapeutic use , Patient Compliance , Adolescent , Blood Glucose Self-Monitoring , Child , Diabetes Mellitus, Type 1/blood , Female , Glycated Hemoglobin/analysis , Humans , Hypoglycemic Agents/administration & dosage , Insulin/administration & dosage , Male , Young Adult
9.
Proc Natl Acad Sci U S A ; 113(13): E1826-34, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26976580

ABSTRACT

Minimally invasive detection of cell death could prove an invaluable resource in many physiologic and pathologic situations. Cell-free circulating DNA (cfDNA) released from dying cells is emerging as a diagnostic tool for monitoring cancer dynamics and graft failure. However, existing methods rely on differences in DNA sequences in source tissues, so that cell death cannot be identified in tissues with a normal genome. We developed a method of detecting tissue-specific cell death in humans based on tissue-specific methylation patterns in cfDNA. We interrogated tissue-specific methylome databases to identify cell type-specific DNA methylation signatures and developed a method to detect these signatures in mixed DNA samples. We isolated cfDNA from plasma or serum of donors, treated the cfDNA with bisulfite, PCR-amplified the cfDNA, and sequenced it to quantify cfDNA carrying the methylation markers of the cell type of interest. Pancreatic ß-cell DNA was identified in the circulation of patients with recently diagnosed type-1 diabetes and islet-graft recipients; oligodendrocyte DNA was identified in patients with relapsing multiple sclerosis; neuronal/glial DNA was identified in patients after traumatic brain injury or cardiac arrest; and exocrine pancreas DNA was identified in patients with pancreatic cancer or pancreatitis. This proof-of-concept study demonstrates that the tissue origins of cfDNA and thus the rate of death of specific cell types can be determined in humans. The approach can be adapted to identify cfDNA derived from any cell type in the body, offering a minimally invasive window for diagnosing and monitoring a broad spectrum of human pathologies as well as providing a better understanding of normal tissue dynamics.


Subject(s)
DNA Methylation , DNA/blood , Insulin-Secreting Cells/pathology , Oligodendroglia/pathology , Adolescent , Adult , Aged , Brain Ischemia/genetics , Brain Ischemia/pathology , Case-Control Studies , Cell Death , Child , Child, Preschool , DNA/metabolism , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Female , Genetic Markers , Humans , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/genetics , Multiple Sclerosis, Relapsing-Remitting/pathology , Organ Specificity , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatitis, Chronic/genetics , Pancreatitis, Chronic/pathology , Promoter Regions, Genetic , Sensitivity and Specificity , Young Adult
10.
Pediatr Endocrinol Rev ; 16(3): 359-368, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30888126

ABSTRACT

Due to well-designed studies of birth cohorts and at-risk individuals, our understanding of the natural history of pre- and early type 1 diabetes (T1D) has advanced considerably over the past decade. Genetic risk scores can predict with increasing precision and accuracy who is at risk for T1D, and early staging based upon islet autoantibody status allows for improved mechanistic and natural history studies as well as improved clinical trial design. A growing number of children are being diagnosed with islet autoimmunity prior to the onset of symptoms, and confusion remains surrounding their proper management. These patients should have access to appropriate counseling and should be referred to a center that can provide information regarding current prevention trials. In the future, a successful prevention strategy for T1D would justify population-based screening for all children.


Subject(s)
Diabetes Mellitus, Type 1 , Autoantibodies , Autoimmunity , Child , Humans , Mass Screening , Risk Factors
11.
Diabetologia ; 61(4): 954-958, 2018 04.
Article in English | MEDLINE | ID: mdl-29128936

ABSTRACT

AIMS/HYPOTHESIS: Diabetes research studies routinely rely upon the use of tissue samples from human organ donors. It remains unclear whether the length of hospital stay prior to organ donation affects the presence of cells infiltrating the pancreas or the frequency of replicating beta cells. METHODS: To address this, 39 organ donors without diabetes were matched for age, sex, BMI and ethnicity in groups of three. Within each group, donors varied by length of hospital stay immediately prior to organ donation (<3 days, 3 to <6 days, or ≥6 days). Serial sections from tissue blocks in the pancreas head, body and tail regions were immunohistochemically double stained for insulin and CD45, CD68, or Ki67. Slides were electronically scanned and quantitatively analysed for cell positivity. RESULTS: No differences in CD45+, CD68+, insulin+, Ki67+ or Ki67+/insulin+ cell frequencies were found when donors were grouped according to duration of hospital stay. Likewise, no interactions were observed between hospitalisation group and pancreas region, age, or both; however, with Ki67 staining, cell frequencies were greater in the body vs the tail region of the pancreas (∆ 0.65 [unadjusted 95% CI 0.25, 1.04]; p = 0.002) from donors <12 year of age. Interestingly, frequencies were less in the body vs tail region of the pancreas for both CD45+ cells (∆ -0.91 [95% CI -1.71, -0.10]; p = 0.024) and insulin+ cells (∆ -0.72 [95% CI -1.10, -0.34]; p < 0.001). CONCLUSIONS/INTERPRETATION: This study suggests that immune or replicating beta cell frequencies are not affected by the length of hospital stay prior to donor death in pancreases used for research. DATA AVAILABILITY: All referenced macros (adopted and developed), calculations, programming code and numerical dataset files (including individual-level donor data) are freely available on GitHub through Zenodo at https://doi.org/10.5281/zenodo.1034422.


Subject(s)
Hospitalization , Length of Stay , Pancreas Transplantation , Pancreas/pathology , Adolescent , Body Mass Index , Child , Death , Diabetes Mellitus/pathology , Female , Humans , Immunohistochemistry , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Male , Tissue Donors , Tissue and Organ Procurement , Treatment Outcome , Young Adult
12.
JAMA ; 318(19): 1891-1902, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29164254

ABSTRACT

Importance: Type 1 diabetes requires major lifestyle changes and carries increased morbidity and mortality. Prevention or delay of diabetes would have major clinical effect. Objective: To determine whether oral insulin delays onset of type 1 diabetes in autoantibody-positive relatives of patients with type 1 diabetes. Design, Setting, and Participants: Between March 2, 2007, and December 21, 2015, relatives with at least 2 autoantibodies, including insulin autoantibodies and normal glucose tolerance, were enrolled in Canada, the United States, Australia, New Zealand, the United Kingdom, Italy, Sweden, Finland, and Germany. The main study group (n = 389) had first-phase insulin release on an intravenous glucose tolerance test that was higher than the threshold. The 55 patients in the secondary stratum 1 had an identical antibody profile as the main study group except they had first-phase insulin release that was lower than the threshold. Secondary strata 2 (n = 114) and strata 3 (n = 3) had different autoantibody profiles and first-phase insulin release threshold combinations. Follow-up continued through December 31, 2016. Interventions: Randomization to receive 7.5 mg/d of oral insulin (n = 283) or placebo (n = 277), including participants in the main study group who received oral insulin (n = 203) or placebo (n = 186). Main Outcome and Measures: The primary outcome was time to diabetes in the main study group. Significance was based on a 1-sided threshold of .05, and 1-sided 95% CIs are reported. Results: Of a total of 560 randomized participants (median enrollment age, 8.2 years; interquartile range [IQR], 5.7-12.1 years; 170 boys [60%]; 90.7% white non-Hispanic; 57.6% with a sibling with type 1 diabetes), 550 completed the trial including 389 participants (median age, 8.4 years; 245 boys [63%]), 382 (96%) in the main study group. During a median follow-up of 2.7 years (IQR, 1.5-4.6 years) in the main study group, diabetes was diagnosed in 58 participants (28.5%) in the oral insulin group and 62 (33%) in the placebo group. Time to diabetes was not significantly different between the 2 groups (hazard ratio [HR], 0.87; 95% CI, 0-1.2; P = .21). In secondary stratum 1 (n = 55), diabetes was diagnosed in 13 participants (48.1%) in the oral insulin group and in 19 participants (70.3%) in the placebo group. The time to diabetes was significantly longer with oral insulin (HR, 0.45; 95% CI, 0-0.82; P = .006). The HR for time to diabetes for the between-group comparisons for the 116 participants in the other secondary stratum was 1.03 (95% CI, 0-2.11; P = .53) and for the entire cohort of 560 participants was 0.83 (95% CI, 0-1.07; P = .11), which were not significantly different. The most common adverse event was infection (n = 254), with 134 events in the oral insulin group and 120 events in the placebo group, but no significant study-related adverse events occurred. Conclusions and Relevance: Among autoantibody-positive relatives of patients with type 1 diabetes, oral insulin at a dose of 7.5 mg/d, compared with placebo, did not delay or prevent the development of type 1 diabetes over 2.7 years. These findings do not support oral insulin as used in this study for diabetes prevention. Trial Registration: clinicaltrials.gov Identifier: NCT00419562.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Administration, Oral , Adolescent , Autoantibodies/blood , Child , Child, Preschool , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/genetics , Family , Female , Follow-Up Studies , Glucose Tolerance Test , Humans , Hypoglycemic Agents/adverse effects , Insulin/adverse effects , Insulin/immunology , Male , Treatment Failure
13.
Diabetologia ; 59(1): 217-221, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26358584

ABSTRACT

AIMS/HYPOTHESIS: Previous studies of pancreases obtained at autopsy or by radiography note reduced pancreas weight (PW) and size, respectively, in type 1 diabetes; this finding is widely considered to be the result of chronic insulinopenia. This literature is, however, limited with respect to the influence of age, sex, anthropometric factors and disease duration on these observations. Moreover, data are sparse for young children, a group of particular interest for type 1 diabetes. We hypothesised that the pancreas-to-body weight ratio would normalise confounding inter-subject factors, thereby permitting better characterisation of PW in type 1 diabetes. METHODS: Transplant-grade pancreases were recovered from 216 organ donors with type 1 diabetes (n = 90), type 2 diabetes (n = 40) and no diabetes (n = 86). Whole-organ and head, body and tail weights were determined. The relative PW (RPW; PW [g] / body weight [kg]) was calculated and tested for normalisation of potential differences due to age, sex and BMI. RESULTS: PW significantly correlated with body weight in control donors (R (2) = 0.76, p < 0.001) while RPW (1.03 ± 0.36, mean ± SD) did not significantly differ across ages (0-58 years). Donors with type 1 diabetes (0.57 ± 0.18, p < 0.001), but not those with type 2 diabetes (0.93 ± 0.30), had significantly lower RPW. The relative weights of each pancreatic region from donors with type 1 diabetes were significantly smaller than those of regions from control donors and donors with type 2 diabetes (p < 0.001). Perhaps most interestingly, the RPW was not significantly associated with duration of type 1 diabetes or type 2 diabetes. CONCLUSIONS/INTERPRETATION: RPW allows for comparisons across a wide range of donor ages by eliminating confounding variables. These data validate an interesting feature of the type 1 diabetes pancreas and underscore the need for additional studies to identify the mechanistic basis for this finding, including those beyond the chronic loss of endogenous insulin secretion.


Subject(s)
Diabetes Mellitus, Type 1/physiopathology , Pancreas/physiopathology , Adolescent , Adult , Age Factors , Anthropometry , Autopsy , Body Mass Index , Body Weight , Child , Child, Preschool , Diabetes Mellitus, Type 2/physiopathology , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Organ Size , Young Adult
14.
Pediatr Diabetes ; 17(5): 327-36, 2016 08.
Article in English | MEDLINE | ID: mdl-26153338

ABSTRACT

OBJECTIVE: Hemoglobin A1c (HbA1c) levels among individuals with type 1 diabetes (T1D) influence the longitudinal risk for diabetes-related complications. Few studies have examined HbA1c trends across time in children, adolescents, and young adults with T1D. This study examines changes in glycemic control across the specific transition periods of pre-adolescence-to-adolescence and adolescence-to-young adulthood, and the demographic and clinical factors associated with these changes. RESEARCH DESIGN AND METHODS: Available HbA1c lab results for up to 10 yr were collected from medical records at 67 T1D Exchange clinics. Two retrospective cohorts were evaluated: the pre-adolescent-to-adolescent cohort consisting of 85 016 HbA1c measurements from 6574 participants collected when the participants were 8-18 yr old and the adolescent-to-young adult cohort, 2200 participants who were 16-26 yr old at the time of 17 279 HbA1c measurements. RESULTS: HbA1c in the 8-18 cohort increased over time after age 10 yr until ages 16-17; followed by a plateau. HbA1c levels in the 16-26 cohort remained steady from 16-18, and then gradually declined. For both cohorts, race/ethnicity, income, health insurance, and pump use were all significant in explaining individual variations in age-centered HbA1c (p < 0.001). For the 8-18 cohort, insulin pump use, age of onset, and health insurance were significant in predicting individual HbA1c trajectory. CONCLUSIONS: Glycemic control among patients 8-18 yr old worsens over time, through age 16. Elevated HbA1c levels observed in 18 yr-olds begin a steady improvement into early adulthood. Focused interventions to prevent deterioration in glucose control in pre-adolescence, adolescence, and early adulthood are needed.


Subject(s)
Diabetes Mellitus, Type 1/blood , Glycated Hemoglobin/metabolism , Registries , Age of Onset , Blood Glucose , Child , Child, Preschool , Diabetes Mellitus, Type 1/drug therapy , Female , Humans , Hypoglycemic Agents/administration & dosage , Insulin/administration & dosage , Insurance Coverage , Longitudinal Studies , Male , Retrospective Studies , Sex Factors , Socioeconomic Factors
15.
Diabetologia ; 58(5): 980-7, 2015 May.
Article in English | MEDLINE | ID: mdl-25660258

ABSTRACT

AIMS/HYPOTHESIS: Islet autoantibodies, in addition to elevated blood glucose, define type 1 diabetes. These autoantibodies are detectable for a variable period of time before diabetes onset. Thus, the occurrence of islet autoantibodies is associated with the beginning of the disease process. The age at, and order in, which autoantibodies appear may be associated with different genetic backgrounds or environmental exposures, or both. METHODS: Infants with HLA-DR high-risk genotypes (DR3/4, DR4/4, DR4/8 and DR3/3) were enrolled and prospectively followed with standardised autoantibody assessments quarterly throughout the first 4 years of life and then semi-annually thereafter. RESULTS: Autoantibodies appeared in 549/8,503 (6.5%) children during 34,091 person-years of follow-up. Autoantibodies at 3 (0.1%) and 6 (0.2%) months of age were rare. Of the 549, 43.7% had islet autoantibodies to insulin (IAA) only, 37.7% had glutamic acid decarboxylase autoantibodies (GADA) only, 13.8% had both GADA and IAA only, 1.6% had insulinoma antigen-2 only and 3.1% had other combinations. The incidence of IAA only peaked within the first year of life and declined over the following 5 years, but GADA only increased until the second year and remained relatively constant. GADA only were more common than IAA only in HLA-DR3/3 children but less common in HLA-DR4/8 children. CONCLUSIONS/INTERPRETATION: Islet autoantibodies can occur very early in life and the order of appearance was related to HLA-DR-DQ genotype.


Subject(s)
Autoantibodies/blood , Diabetes Mellitus, Type 1/immunology , Genetic Predisposition to Disease , HLA-DQ Antigens/genetics , Islets of Langerhans/immunology , Child , Child, Preschool , Diabetes Mellitus, Type 1/genetics , Female , Genotype , Humans , Insulin/genetics , Male
16.
Pediatr Diabetes ; 16(8): 565-72, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26392298

ABSTRACT

BACKGROUND: Children at high risk for developing type 1 diabetes (T1D) can be identified on the basis of human leukocyte antigen (HLA) genotype and the subsequent development of islet cell autoantibodies. Several studies have documented reduced incidence of diabetic ketoacidosis (DKA) in new-onset T1D when high-risk children are identified at an early age. Many have questioned whether general population screening for T1D risk should be standard of practice. We sought to perform a purely economic, cost-benefit analysis to determine if a screening program to reduce the incidence of DKA at diagnosis in children less than 5 yr is cost effective. METHODS: We compared the cost of population screening with the benefit of preventing DKA. The cost of screening included one-time HLA typing on the entire population followed by islet cell autoantibody testing in high-risk children every 6 months until age 5 yr. The potential benefits of screening included reductions in parental lost income, medical expenses, morbidity, and mortality. RESULTS: Screening for T1D risk for the sole purpose of reducing the cost of DKA at onset of T1D was not economically viable unless HLA testing and autoantibody testing could be performed for less than $1 and $0.03, respectively. CONCLUSIONS: Current screening costs far outweigh the economic benefits of preventing new-onset DKA in children under 5 yr of age.


Subject(s)
Diabetes Mellitus, Type 1/diagnosis , Diabetic Ketoacidosis/prevention & control , Mass Screening/economics , Child, Preschool , Cost-Benefit Analysis , Diabetic Ketoacidosis/economics , Humans
17.
Diabetes Spectr ; 28(2): 92-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25987807

ABSTRACT

Patient-centered approaches to disease management are consistently recognized as valuable tools for improving health outcomes, yet studies are rarely designed to elicit adolescent perspectives. This study sought to better understand the perspectives of youths with type 1 diabetes according to key demographic variables. We conducted an exploratory study through which 40 youths were provided with disposable cameras and prompted to take five photographs each that captured what diabetes meant to them and to provide narratives to accompany their photo choices. Demographic variables examined included sex, age, disease duration, socioeconomic status (SES), race, and glycemic control (A1C). Content analysis was used for photos and open-ended responses to assign photo index scores, which were then analyzed by demographic variables using Mann-Whitney U tests for statistical significance. Analysis of photos/narratives (n = 202) revealed five main types of representations depicted by at least 50% of the young people. "Challenge" photos included diabetes supplies as tethering, food as a source of frustration, and the body as a territory for disease encroachment. "Resilience" photos included coping mechanisms and symbols of resistance. Overall, these representations were consistent across demographic categories with two exceptions. Males took more food depictions than females (P <0.005) and had fewer coping depictions (P <0.05). Youths from more affluent households were more likely to take photos of resistance (P <0.05). The use of photo index scores expands previous studies using photography by comparing demographic variation within a sample. Our findings provide insight into coping strategies and indicate that SES may provide an advantage for affluent youths in meeting diabetes-specific challenges.

18.
Clin Diabetes ; 33(3): 159, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26202839

ABSTRACT

[This corrects the article on p. 62 in vol. 33, PMID: 25897185.].

19.
Clin Diabetes ; 33(2): 62-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25897185

ABSTRACT

IN BRIEF Low socioeconomic status (SES) is consistently identified as a major risk factor for poor health outcomes in youths with type 1 diabetes, yet little is known about the social factors that yield such disparities. This study used survey research to examine the role of SES by focusing on differential resourcing in social support systems for youths with type 1 diabetes and their parents/caregivers. We identified significant inequalities in social support systems and found that parents from lower-income households engage in few coping activities and rarely identify a primary care provider as the main point of contact when facing a diabetes-related problem. Our findings underscore the need to better connect low SES families to diabetes-specific professional resourcing and to raise awareness about the importance of extracurricular activities as a form of social support for youths.

20.
Curr Opin Pediatr ; 26(4): 493-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24905103

ABSTRACT

PURPOSE OF REVIEW: This review discusses the IgG4-related disease spectrum (IgG4-RD), the autoimmune polyglandular syndromes (APS), the association of IgG4 with APS, and possible pathobiology. RECENT FINDINGS: IgG4-RD is a multiorgan autoimmune disorder characterized by fibrous inflammation, IgG4-positive plasma cell infiltration in affected tissues, and elevated serum concentrations of IgG4. IgG4-RD can affect any organ and has a heterogeneous presentation. Consensus criteria for diagnosis in specific organs have been established. The recognition and diagnosis of IgG4-RD are crucial because the disease responds favorably to immunosuppression (e.g., glucocorticoids, rituximab). The precise mechanisms leading to disease are unknown, but IgG4 antibodies may undergo a half antibody exchange, which renders them incapable of activating the complement pathway. SUMMARY: Despite significant advances in disease recognition and treatment strategies, the disorder remains poorly understood. The precise role of IgG4, whether it is protective or pathogenic, is still being debated.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/therapeutic use , Glucocorticoids/therapeutic use , Immunoglobulin G/immunology , Immunologic Factors/therapeutic use , Immunosuppressive Agents/therapeutic use , Inflammation/immunology , Polyendocrinopathies, Autoimmune/immunology , Autoantibodies/blood , Humans , Immunoglobulin G/drug effects , Inflammation/drug therapy , Polyendocrinopathies, Autoimmune/drug therapy , Polyendocrinopathies, Autoimmune/physiopathology , Rituximab
SELECTION OF CITATIONS
SEARCH DETAIL