Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38712386

ABSTRACT

BACKGROUND: Essentially all individuals with multiple autoantibodies will develop clinical type 1 diabetes. Multiple AABs and normal glucose tolerance define Stage 1 diabetes; abnormal glucose tolerance defines Stage 2. However, the rate of progression within these stages is heterogeneous, necessitating personalized risk calculators to improve clinical implementation. METHODS: We developed 3 models using TrialNet's Pathway to Prevention data to accommodate the reality that not all risk variables are clinically available. The Small model included AAB status, fasting glucose, HbA1c and age, while the Medium and Large models added predictors of disease progression measured via oral glucose tolerance testing. FINDINGS: All models markedly improved granularity regarding personalized risk missing from current categories of stages of T1D. Model derived risk calculations are consistent with the expected reduction of risk with increasing age and increase in risk with higher glucose and lower insulin secretion, illustrating the suitability of the models. Adding glucose and insulin secretion data altered model predicted probabilities within Stages. In those with high 2-hour glucose, a high C-peptide markedly decreased predicted risk; lower C-peptide obviated the age-dependent risk of 2-hour glucose alone, providing a more nuanced estimate of rate of disease progression within Stage 2. CONCLUSIONS: While essentially all those with multiple AABs will develop type 1 diabetes, the rate of progression is heterogeneous and not explained by any individual single risk variable. The model-based probabilities developed here provide an adaptable personalized risk calculator to better inform decisions about how and when to monitor disease progression in clinical practice.

2.
Sci Transl Med ; 16(746): eadn2404, 2024 05 08.
Article in English | MEDLINE | ID: mdl-38718135

ABSTRACT

CD4+CD25hiCD127lo/-FOXP3+ regulatory T cells (Tregs) play a key role in preventing autoimmunity. In autoimmune type 1 diabetes (T1D), adoptive transfer of autologous polyclonal Tregs has been shown to be safe in adults in phase 1 clinical trials. We explored factors contributing to efficacy of autologous polyclonal expanded Tregs (expTregs) in a randomized phase 2 multi-center, double-blind, clinical trial (Sanford/Lisata Therapeutics T-Rex phase 2 trial, ClinicalTrials.gov NCT02691247). One hundred ten treated children and adolescents with new-onset T1D were randomized 1:1:1 to high-dose (20 × 106 cells/kilogram) or low-dose (1 × 106 cells/kilogram) treatments or to matching placebo. Cytometry as well as bulk and single-cell RNA sequencing were performed on selected expTregs and peripheral blood samples from participants. The single doses of expTregs were safe but did not prevent decline in residual ß cell function over 1 year compared to placebo (P = 0.94 low dose, P = 0.21 high dose), regardless of age or baseline C-peptide. ExpTregs were highly activated and suppressive in vitro. A transient increase of activated memory Tregs was detectable 1 week after infusion in the high-dose cohort, suggesting effective transfer of expTregs. However, the in vitro fold expansion of expTregs varied across participants, even when accounting for age, and lower fold expansion and its associated gene signature were linked with better C-peptide preservation regardless of Treg dose. These results suggest that a single dose of polyclonal expTregs does not alter progression in T1D; instead, Treg quality may be an important factor.


Subject(s)
Diabetes Mellitus, Type 1 , T-Lymphocytes, Regulatory , Humans , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , T-Lymphocytes, Regulatory/immunology , Child , Adolescent , Male , Female , Double-Blind Method , Child, Preschool , Transplantation, Autologous
3.
J Autoimmun ; 143: 103161, 2024 02.
Article in English | MEDLINE | ID: mdl-38141419

ABSTRACT

Although type 1 diabetes (T1D) results from the autoimmune destruction of the insulin-producing ß-cells, its treatment is largely restricted to exogenous insulin administration. Only few therapies targeting the autoaggressive immune system have been introduced into clinical practice or are considered in clinical trials. Here, we provide a gene expression profile of the islet microenvironment obtained by laser-dissection microscopy in an inducible mouse model. Thereby, we have identified novel targets for immune intervention. Increased gene expression of most inflammatory proteins was apparent at day 10 after T1D induction and largely paralleled the observed degree of insulitis. We further focused on genes involved in leukocyte migration, including chemokines and their receptors. Besides the critical chemokine CXCL10, we found several other chemokines upregulated locally in temporary or chronic manner. Localization of the chemokine ligand/receptor pairs to the islet microenvironment has been confirmed by RNAscope. Interference with the CXCL16-CXCR6 and CX3CL1-CX3CR1 axes, but not the CCL5-CCR1/3/5 axis, resulted in reduced insulitis and lower T1D incidence. Further, we found that the receptors for the differentially expressed chemokines CXCL10, CXCL16 and CX3CL1 are distributed unevenly among islet autoantigen-specific T cells, which explains why the interference with just one chemokine axis cannot completely abrogate insulitis and T1D.


Subject(s)
Diabetes Mellitus, Type 1 , Islets of Langerhans , Mice , Animals , Mice, Inbred NOD , Chemokine CXCL10/genetics , Insulin/metabolism
4.
Glob Chang Biol ; 29(24): 6900-6911, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37804212

ABSTRACT

The global decline of terrestrial species is largely due to the degradation, loss and fragmentation of their habitats. The conversion of natural ecosystems for cropland, rangeland, forest products and human infrastructure are the primary causes of habitat deterioration. Due to the paucity of data on the past distribution of species and the scarcity of fine-scale habitat conversion maps, however, accurate assessment of the recent effects of habitat degradation, loss and fragmentation on the range of mammals has been near impossible. We aim to assess the proportions of available habitat within the lost and retained parts of mammals' distribution ranges, and to identify the drivers of habitat availability. We produced distribution maps for 475 terrestrial mammals for the range they occupied 50 years ago and compared them to current range maps. We then calculated the differences in the percentage of 'area of habitat' (habitat available to a species within its range) between the lost and retained range areas. Finally, we ran generalized linear mixed models to identify which variables were more influential in determining habitat availability in the lost and retained parts of the distribution ranges. We found that 59% of species had a lower proportion of available habitat in the lost range compared to the retained range, thus hypothesizing that habitat loss could have contributed to range declines. The most important factors negatively affecting habitat availability were the conversion of land to rangeland and high density of livestock. Significant intrinsic traits were those related to reproductive timing and output, habitat breadth and medium body size. Our findings emphasize the importance of implementing conservation strategies to mitigate the impacts caused by human activities on the habitats of mammals, and offer evidence indicating which species have the potential to reoccupy portions of their former range if other threats cease to occur.


Subject(s)
Ecosystem , Livestock , Animals , Humans , Conservation of Natural Resources , Mammals , Forests
5.
J Clin Endocrinol Metab ; 109(1): 57-67, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-37572381

ABSTRACT

CONTEXT: The value of continuous glucose monitoring (CGM) for monitoring autoantibody (AAB)-positive individuals in clinical trials for progression of type 1 diabetes (T1D) is unknown. OBJECTIVE: Compare CGM with oral glucose tolerance test (OGTT)-based metrics in prediction of T1D. METHODS: At academic centers, OGTT and CGM data from multiple-AAB relatives were evaluated for associations with T1D diagnosis. Participants were multiple-AAB-positive individuals in a TrialNet Pathway to Prevention (TN01) CGM ancillary study (n = 93). The intervention was CGM for 1 week at baseline, 6 months, and 12 months. Receiver operating characteristic (ROC) curves of CGM and OGTT metrics for prediction of T1D were analyzed. RESULTS: Five of 7 OGTT metrics and 29/48 CGM metrics but not HbA1c differed between those who subsequently did or did not develop T1D. ROC area under the curve (AUC) of individual CGM values ranged from 50% to 69% and increased when adjusted for age and AABs. However, the highest-ranking metrics were derived from OGTT: 4/7 with AUC ∼80%. Compared with adjusted multivariable models using CGM data, OGTT-derived variables, Index60 and DPTRS (Diabetes Prevention Trial-Type 1 Risk Score), had higher discriminative ability (higher ROC AUC and positive predictive value with similar negative predictive value). CONCLUSION: Every 6-month CGM measures in multiple-AAB-positive individuals are predictive of subsequent T1D, but less so than OGTT-derived variables. CGM may have feasibility advantages and be useful in some settings. However, our data suggest there is insufficient evidence to replace OGTT measures with CGM in the context of clinical trials.


Subject(s)
Diabetes Mellitus, Type 1 , Humans , Diabetes Mellitus, Type 1/diagnosis , Glucose Tolerance Test , Blood Glucose/metabolism , Autoantibodies , Blood Glucose Self-Monitoring , Continuous Glucose Monitoring
6.
J Clin Endocrinol Metab ; 108(3): 688-696, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36227635

ABSTRACT

CONTEXT: The presence of islet autoimmunity identifies individuals likely to progress to clinical type 1 diabetes (T1D). In clinical research studies, autoantibody screening followed by regular metabolic monitoring every 6 months reduces incidence of diabetic ketoacidosis (DKA) at diagnosis. OBJECTIVE: We hypothesized that DKA reduction can be achieved on a population basis with a reduced frequency of metabolic monitoring visits. We reasoned that prolonged time between the development of T1D and the time of clinical diagnosis ("undiagnosed time") would more commonly result in DKA and thus that limiting undiagnosed time would decrease DKA. METHODS: An analysis was conducted of data from TrialNet's Pathway to Prevention (PTP), a cross-sectional longitudinal study that identifies and follows at-risk relatives of people with T1D. PTP is a population-based study enrolling across multiple countries. A total of 6193 autoantibody (AAB)-positive individuals participated in PTP from March 2004 to April 2019. We developed models of progression to clinical diagnosis for pediatric and adult populations with single or multiple AAB, and summarized results using estimated hazard rate. An optimal monitoring visit schedule was determined for each model to achieve a minimum average level of undiagnosed time for each population. RESULTS: Halving the number of monitoring visits usually conducted in research studies is likely to substantially lower the population incidence of DKA at diagnosis of T1D. CONCLUSION: Our study has clinical implications for the metabolic monitoring of at-risk individuals. Fewer monitoring visits would reduce the clinical burden, suggesting a path toward transitioning monitoring beyond the research setting.


Subject(s)
Diabetes Mellitus, Type 1 , Diabetic Ketoacidosis , Adult , Child , Humans , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/epidemiology , Diabetes Mellitus, Type 1/complications , Diabetic Ketoacidosis/diagnosis , Diabetic Ketoacidosis/epidemiology , Diabetic Ketoacidosis/etiology , Autoantibodies , Cross-Sectional Studies , Longitudinal Studies
7.
Wellcome Open Res ; 8: 514, 2023.
Article in English | MEDLINE | ID: mdl-38911281

ABSTRACT

We present a genome assembly from an individual male Muscardinus avellanarius (the hazel dormouse; Chordata; Mammalia; Rodentia; Gliridae). The genome sequence is 2,497.5 megabases in span. Most of the assembly is scaffolded into 24 chromosomal pseudomolecules, including the X and Y sex chromosomes. The mitochondrial genome has also been assembled and is 16.73 kilobases in length.

8.
Front Immunol ; 12: 683091, 2021.
Article in English | MEDLINE | ID: mdl-34220832

ABSTRACT

In human type 1 diabetes and animal models of the disease, a diverse assortment of immune cells infiltrates the pancreatic islets. CD8+ T cells are well represented within infiltrates and HLA multimer staining of pancreas sections provides clear evidence that islet epitope reactive T cells are present within autoimmune lesions. These bona fide effectors have been a key research focus because these cells represent an intellectually attractive culprit for ß cell destruction. However, T cell receptors are highly diverse in human insulitis. This suggests correspondingly broad antigen specificity, which includes a majority of T cells for which there is no evidence of islet-specific reactivity. The presence of "non-cognate" T cells in insulitis raises suspicion that their role could be beyond that of an innocent bystander. In this perspective, we consider the potential pathogenic contribution of non-islet-reactive T cells. Our intellectual framework will be that of a criminal investigation. Having arraigned islet-specific CD8+ T cells for the murder of pancreatic ß cells, we then turn our attention to the non-target immune cells present in human insulitis and consider the possible regulatory, benign, or effector roles that they may play in disease. Considering available evidence, we overview the case that can be made that non-islet-reactive infiltrating T cells should be suspected as co-conspirators or accessories to the crime and suggest some possible routes forward for reaching a better understanding of their role in disease.


Subject(s)
Autoimmunity/immunology , Diabetes Mellitus, Type 1/etiology , Disease Susceptibility , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Autoimmunity/genetics , Biomarkers , Cell Communication/genetics , Cell Communication/immunology , Cellular Microenvironment/immunology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Humans , Islets of Langerhans/pathology , T-Lymphocyte Subsets/pathology
9.
Ecol Appl ; 31(2): e02272, 2021 03.
Article in English | MEDLINE | ID: mdl-33290622

ABSTRACT

Natural resources such as waterbodies, public parks, and wildlife refuges attract people from varying distances on the landscape, creating "social-ecological catchments." Catchments have provided great utility for understanding physical and social relationships within specific disciplines. Yet, catchments are rarely used across disciplines, such as its application to understand complex spatiotemporal dynamics between mobile human users and patchily distributed natural resources. We collected residence ZIP codes from 19,983 angler parties during 2014-2017 to construct seven angler-waterbody catchments in Nebraska, USA. We predicted that sizes of dense (10% utilization distribution) and dispersed (95% utilization distribution) angler-waterbody catchments would change across seasons and years as a function of diverse resource selection among mobile anglers. Contrary to expectations, we revealed that catchment size was invariant. We discuss how social (conservation actions) and ecological (low water quality, reduction in species diversity) conditions are expected to impact landscape patterns in resource use. We highlight how this simple concept and user-friendly technique can inform timely landscape-level conservation decisions within coupled social-ecological systems that are currently difficult to study and understand.


Subject(s)
Conservation of Natural Resources , Fisheries , Ecosystem , Humans , Nebraska
10.
Sci Adv ; 6(42)2020 10.
Article in English | MEDLINE | ID: mdl-33067232

ABSTRACT

Preproinsulin (PPI) is presumably a crucial islet autoantigen found in patients with type 1 diabetes (T1D) but is also recognized by CD8+ T cells from healthy individuals. We quantified PPI-specific CD8+ T cells within different areas of the human pancreas from nondiabetic controls, autoantibody-positive donors, and donors with T1D to investigate their role in diabetes development. This spatial cellular quantitation revealed unusually high frequencies of autoreactive CD8+ T cells supporting the hypothesis that PPI is indeed a key autoantigen. To our surprise, PPI-specific CD8+ T cells were already abundantly present in the nondiabetic pancreas, thus questioning the dogma that T1D is caused by defective thymic deletion or systemic immune dysregulation. During T1D development, these cells accumulated in and around islets, indicating that an islet-specific trigger such as up-regulation of major histocompatibility complex class I might be essential to unmask beta cells to the immune system.


Subject(s)
Diabetes Mellitus, Type 1 , Pancreas, Exocrine , Autoantigens , CD8-Positive T-Lymphocytes , Humans , Insulin , Protein Precursors
11.
Front Endocrinol (Lausanne) ; 11: 606434, 2020.
Article in English | MEDLINE | ID: mdl-33469446

ABSTRACT

Since the establishment of the network for pancreatic organ donors with diabetes (nPOD), we have gained unprecedented insight into the pathology of human type 1 diabetes. Many of the pre-existing "dogmas", mostly derived from studies of animal models and sometimes limited human samples, have to be revised now. For example, we have learned that autoreactive CD8 T cells are present even in healthy individuals within the exocrine pancreas. Furthermore, their "attraction" to islets probably relies on beta-cell intrinsic events, such as the over-expression of MHC class I and resulting presentation of autoantigens such as (prepro)insulin. In addition, we are discovering other signs of beta-cell dysfunction, possibly at least in part due to stress, such as the over-expression of certain cytokines. This review summarizes the latest developments focusing on cytokines and autoreactive CD8 T cells in human type 1 diabetes pathogenesis.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytokines , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Animals , Diabetes Mellitus, Type 1/pathology , Humans , Pancreas/immunology , Pancreas/pathology
12.
Diabetes ; 66(1): 113-126, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27797910

ABSTRACT

Type 1 diabetes (T1D) results from the autoimmune destruction of insulin-producing ß-cells in the pancreas. Thereby, the chemokine CXC-motif ligand 10 (CXCL10) plays an important role in the recruitment of autoaggressive lymphocytes to the islets of Langerhans. Transplantation of isolated islets as a promising therapy for T1D has been hampered by early graft rejection. Here, we investigated the influence of CXCL10 on the autoimmune destruction of islet isografts using RIP-LCMV mice expressing a lymphocytic choriomeningitis virus (LCMV) protein in the ß-cells. RIP-LCMV islets express CXCL10 after isolation and maintain CXCL10 production after engraftment. Thus, we isolated islets from either normal or CXCL10-deficient RIP-LCMV mice and transferred them under the kidney capsule of diabetic RIP-LCMV mice. We found that the autoimmune destruction of CXCL10-deficient islet isografts was significantly reduced. The autoimmune destruction was also diminished in mice administered with an anti-CXCL10 antibody. The persistent protection from autoimmune destruction was paralleled by an increase in FoxP3+ regulatory T cells within the cellular infiltrates around the islet isografts. Consequently, CXCL10 might influence the cellular composition locally in the islet graft, thereby playing a role in the autoimmune destruction. CXCL10 might therefore constitute a potential therapeutic target to prolong islet graft survival.


Subject(s)
Chemokine CXCL10/metabolism , Diabetes Mellitus, Type 1/metabolism , Islets of Langerhans/metabolism , Isografts/metabolism , Animals , Chemokine CXCL10/genetics , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Graft Rejection/genetics , Graft Survival/genetics , Graft Survival/physiology , Immunohistochemistry , Islets of Langerhans Transplantation , Lymphocytic choriomeningitis virus/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains
13.
Front Pharmacol ; 6: 317, 2015.
Article in English | MEDLINE | ID: mdl-26793108

ABSTRACT

Interleukin (IL)-22 is a cytokine displaying tissue protective and pro-regenerative functions in various preclinical disease models. Anti-bacterial, pro-proliferative, and anti-apoptotic properties mediated by activation of the transcription factor signal transducer and activator of transcription (STAT)-3 are key to biological functions of this IL-10 family member. Herein, we introduce RINm5F insulinoma cells as rat ß-cell line that, under the influence of IL-22, displays activation of STAT3 with induction of its downstream gene targets Socs3, Bcl3, and Reg3b. In addition, IL-22 also activates STAT1 in this cell type. To refine those observations, IL-22 biological activity was evaluated using ex vivo cultivated murine pancreatic islets. In accord with data on RINm5F cells, islet exposure to IL-22 activated STAT3 and upregulation of STAT3-inducible Socs3, Bcl3, and Steap4 was evident under those conditions. As these observations supported the hypothesis that IL-22 may exert protective functions in toxic ß-cell injury, application of IL-22 was investigated in murine multiple-low-dose streptozotocin (STZ)-induced diabetes. For that purpose, recombinant IL-22 was administered thrice either immediately before and at disease onset (at d4, d6, d8) or closely thereafter (at d8, d10, d12). These two IL-22-treatment periods coincide with two early peaks of ß-cell injury detectable in this model. Notably, none of the two IL-22-treatment strategies affected diabetes incidence or blood glucose levels in STZ-treated mice. Moreover, pathological changes in islet morphology analyzed 28 days after disease induction were not ameliorated by IL-22 administration. Taken together, despite being active on rat RINm5F insulinoma cells and murine pancreatic islets, recombinant IL-22 fails to protect pancreatic ß-cells in the tested protocols from toxic effects of STZ and thus is unable to ameliorate disease in the widely used model of STZ-induced diabetes.

14.
Diabetologia ; 57(9): 1911-8, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24939430

ABSTRACT

AIMS/HYPOTHESIS: Subtyping GAD autoantibody (GADA) responses using affinity measurement allows the identification of GADA-positive children with a family history of type 1 diabetes who are at risk of developing diabetes. Here, we asked whether GADA affinity is a useful marker to stratify the risk of type 1 diabetes in GADA-positive schoolchildren from the general population. METHODS: GADA affinity was measured by competitive binding experiments with [(125)I]-labelled and unlabelled human 65 kDa isoform of GAD (GAD65) in sera from 97 GADA-positive children identified in the Karlsburg Type 1 Diabetes Risk Study of a general schoolchild population in north-eastern Germany. GADA epitope specificity was determined using radiobinding assays with [(35)S]-labelled GAD65/67 kDa isoform of GAD (GAD67) chimeric proteins. RESULTS: GADA affinity was high, ≥ 10(10) l/mol, in 33 of 35 multiple islet autoantibody-positive children. In contrast, the affinity ranged widely among 62 single GADA-positive children (median 3.1 × 10(9) l/mol; range 5.6 × 10(6) to >4.0 × 10(11) l/mol; p < 0.0001). High-affinity GADA were associated with HLA-DRB1*03 (p = 0.02) and predominantly directed against the C-terminal and/or middle part of the GAD65 protein. At follow-up, the affinity remained relatively constant. Five of the single GADA-positive children developed additional islet autoantibodies and had high-affinity GADA. Twenty-six children progressed to type 1 diabetes; among them, 23 had GADA affinities of ≥ 10(10) l/mol before disease onset. CONCLUSIONS/INTERPRETATION: Schoolchildren from the general population may develop heterogeneous GADA responses, and a high affinity can identify those GADA-positive children who are more likely to progress to type 1 diabetes.


Subject(s)
Autoantibodies/metabolism , Glutamate Decarboxylase/immunology , Adolescent , Child , Diabetes Mellitus, Type 1/immunology , Female , Humans , Male
15.
Curr Opin Rheumatol ; 24(4): 417-23, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22504578

ABSTRACT

PURPOSE OF REVIEW: It is the current opinion that pathogens, such as viruses, are contributing to the development of type 1 diabetes (T1D) in susceptible individuals. This opinion is based on epidemiological associations, direct isolation of pathogens from the islets of Langerhans, as well as a large amount of data from various experimental animal models. Human enteroviruses have dominated the literature associated with the etiology of T1D. However, virus infections have also been reported to protect from autoimmune disorders. RECENT FINDINGS: Here we review the evidence for virus infections to be involved in the pathogenesis of T1D and discuss potential mechanisms of how such infections could accelerate the destruction of insulin-producing ß-cells. In addition, we will review evidence from epidemiologic and experimental animal studies showing that virus infections could also have protective properties. SUMMARY: Virus infections play an important role in the pathogenesis of T1D by inducing or accelerating the autodestructive process, but also by protecting from autoimmunity. Thus, multiple sequential infections might shape the autoreactive immune repertoire and the pathogenesis of T1D in a complex fashion.


Subject(s)
Diabetes Mellitus, Type 1/virology , Virus Diseases/complications , Animals , Diabetes Mellitus, Type 1/epidemiology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Disease Models, Animal , Enterovirus Infections/complications , Enterovirus Infections/epidemiology , Evidence-Based Medicine , Humans , Islets of Langerhans/virology , Molecular Mimicry/immunology , Virus Diseases/epidemiology , Virus Diseases/immunology
16.
Pediatr Nurs ; 31(1): 61; author reply 61-2, 2005.
Article in English | MEDLINE | ID: mdl-15794328
SELECTION OF CITATIONS
SEARCH DETAIL
...