Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.881
Filter
1.
Nat Commun ; 15(1): 3810, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714671

ABSTRACT

Previous studies have revealed heterogeneity in the progression to clinical type 1 diabetes in children who develop islet-specific antibodies either to insulin (IAA) or glutamic acid decarboxylase (GADA) as the first autoantibodies. Here, we test the hypothesis that children who later develop clinical disease have different early immune responses, depending on the type of the first autoantibody to appear (GADA-first or IAA-first). We use mass cytometry for deep immune profiling of peripheral blood mononuclear cell samples longitudinally collected from children who later progressed to clinical disease (IAA-first, GADA-first, ≥2 autoantibodies first groups) and matched for age, sex, and HLA controls who did not, as part of the Type 1 Diabetes Prediction and Prevention study. We identify differences in immune cell composition of children who later develop disease depending on the type of autoantibodies that appear first. Notably, we observe an increase in CD161 expression in natural killer cells of children with ≥2 autoantibodies and validate this in an independent cohort. The results highlight the importance of endotype-specific analyses and are likely to contribute to our understanding of pathogenic mechanisms underlying type 1 diabetes development.


Subject(s)
Autoantibodies , Diabetes Mellitus, Type 1 , Glutamate Decarboxylase , Immunity, Cellular , Humans , Diabetes Mellitus, Type 1/immunology , Autoantibodies/immunology , Autoantibodies/blood , Child , Female , Male , Glutamate Decarboxylase/immunology , Child, Preschool , Adolescent , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/immunology , Insulin/immunology , Islets of Langerhans/immunology , Disease Progression
2.
J Immunol ; 212(12): 1971-1980, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38709159

ABSTRACT

Most pancreatic islets are destroyed immediately after intraportal transplantation by an instant blood-mediated inflammatory reaction (IBMIR) generated through activation of coagulation, complement, and proinflammatory pathways. Thus, effective mitigation of IBMIR may be contingent on the combined use of agents targeting these pathways for modulation. CD47 and thrombomodulin (TM) are two molecules with distinct functions in regulating coagulation and proinflammatory responses. We previously reported that the islet surface can be modified with biotin for transient display of novel forms of these two molecules chimeric with streptavidin (SA), that is, thrombomodulin chimeric with SA (SA-TM) and CD47 chimeric with SA (SA-CD47), as single agents with improved engraftment following intraportal transplantation. This study aimed to test whether islets can be coengineered with SA-TM and SA-CD47 molecules as a combinatorial approach to improve engraftment by inhibiting IBMIR. Mouse islets were effectively coengineered with both molecules without a detectable negative impact on their viability and metabolic function. Coengineered islets were refractory to destruction by IBMIR ex vivo and showed enhanced engraftment and sustained function in a marginal mass syngeneic intraportal transplantation model. Improved engraftment correlated with a reduction in intragraft innate immune infiltrates, particularly neutrophils and M1 macrophages. Moreover, transcripts for various intragraft procoagulatory and proinflammatory agents, including tissue factor, HMGB1 (high-mobility group box-1), IL-1ß, IL-6, TNF-α, IFN-γ, and MIP-1α, were significantly reduced in coengineered islets. These data demonstrate that the transient codisplay of SA-TM and SA-CD47 proteins on the islet surface is a facile and effective platform to modulate procoagulatory and inflammatory responses with implications for both autologous and allogeneic islet transplantation.


Subject(s)
CD47 Antigen , Inflammation , Islets of Langerhans Transplantation , Islets of Langerhans , Mice, Inbred C57BL , Thrombomodulin , Animals , CD47 Antigen/immunology , CD47 Antigen/metabolism , Mice , Islets of Langerhans Transplantation/methods , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Inflammation/immunology , Male , Streptavidin
3.
Front Immunol ; 15: 1375177, 2024.
Article in English | MEDLINE | ID: mdl-38650946

ABSTRACT

Human allogeneic pancreatic islet transplantation is a life-changing treatment for patients with severe Type 1 Diabetes (T1D) who suffer from hypoglycemia unawareness and high risk of severe hypoglycemia. However, intensive immunosuppression is required to prevent immune rejection of the graft, that may in turn lead to undesirable side effects such as toxicity to the islet cells, kidney toxicity, occurrence of opportunistic infections, and malignancies. The shortage of cadaveric human islet donors further limits islet transplantation as a treatment option for widespread adoption. Alternatively, porcine islets have been considered as another source of insulin-secreting cells for transplantation in T1D patients, though xeno-transplants raise concerns over the risk of endogenous retrovirus transmission and immunological incompatibility. As a result, technological advancements have been made to protect transplanted islets from immune rejection and inflammation, ideally in the absence of chronic immunosuppression, to improve the outcomes and accessibility of allogeneic islet cell replacement therapies. These include the use of microencapsulation or macroencapsulation devices designed to provide an immunoprotective environment using a cell-impermeable layer, preventing immune cell attack of the transplanted cells. Other up and coming advancements are based on the use of stem cells as the starting source material for generating islet cells 'on-demand'. These starting stem cell sources include human induced pluripotent stem cells (hiPSCs) that have been genetically engineered to avoid the host immune response, curated HLA-selected donor hiPSCs that can be matched with recipients within a given population, and multipotent stem cells with natural immune privilege properties. These strategies are developed to provide an immune-evasive cell resource for allogeneic cell therapy. This review will summarize the immunological challenges facing islet transplantation and highlight recent bio-engineering and cell-based approaches aimed at avoiding immune rejection, to improve the accessibility of islet cell therapy and enhance treatment outcomes. Better understanding of the different approaches and their limitations can guide future research endeavors towards developing more comprehensive and targeted strategies for creating a more tolerogenic microenvironment, and improve the effectiveness and sustainability of islet transplantation to benefit more patients.


Subject(s)
Diabetes Mellitus, Type 1 , Graft Rejection , Islets of Langerhans Transplantation , Islets of Langerhans Transplantation/methods , Humans , Animals , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Graft Rejection/immunology , Graft Rejection/prevention & control , Biomedical Engineering/methods , Islets of Langerhans/immunology
4.
J Autoimmun ; 146: 103228, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38642507

ABSTRACT

CD6 is a glycoprotein expressed on CD4 and CD8 T cells involved in immunoregulation. CD318 has been identified as a CD6 ligand. The role of CD318 in T cell immunity is restricted as it has only been investigated in a few mice autoimmune models but not in human diseases. CD318 expression was thought to be limited to mesenchymal-epithelial cells and, therefore, contribute to CD6-mediated T cell activation in the CD318-expressing tissue rather than through interaction with antigen-presenting cells. Here, we report CD318 expression in a subpopulation of CD318+ myeloid dendritic (mDC), whereas the other peripheral blood populations were CD318 negative. However, CD318 can be induced by activation: a subset of monocytes treated with LPS and IFNγ and in vitro monocyte derived DCs were CD318+. We also showed that recombinant CD318 inhibited T cell function. Strikingly, CD318+ DCs suppressed the proliferation of autoreactive T cells specific for GAD65, a well-known targeted self-antigen in Type 1 Diabetes (T1D). Our study provides new insight into the role of the CD318/CD6 axis in the immunopathogenesis of inflammation, suggesting a novel immunoregulatory role of CD318 in T cell-mediated autoimmune diseases and identifying a potential novel immune checkpoint inhibitor as a target for intervention in T1D which is an unmet therapeutic need.


Subject(s)
Antigens, CD , Autoantigens , Dendritic Cells , Diabetes Mellitus, Type 1 , Islets of Langerhans , Lymphocyte Activation , Humans , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Lymphocyte Activation/immunology , Autoantigens/immunology , Antigens, CD/metabolism , Antigens, CD/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Antigens, Differentiation, T-Lymphocyte/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Cells, Cultured , Glutamate Decarboxylase
5.
Cell Rep Med ; 5(5): 101535, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38677282

ABSTRACT

Type 1 diabetes (T1D) is a chronic condition in which beta cells are destroyed by immune cells. Despite progress in immunotherapies that could delay T1D onset, early detection of autoimmunity remains challenging. Here, we evaluate the utility of machine learning for early prediction of T1D using single-cell analysis of islets. Using gradient-boosting algorithms, we model changes in gene expression of single cells from pancreatic tissues in T1D and non-diabetic organ donors. We assess if mathematical modeling could predict the likelihood of T1D development in non-diabetic autoantibody-positive donors. While most autoantibody-positive donors are predicted to be non-diabetic, select donors with unique gene signatures are classified as T1D. Our strategy also reveals a shared gene signature in distinct T1D-associated models across cell types, suggesting a common effect of the disease on transcriptional outputs of these cells. Our study establishes a precedent for using machine learning in early detection of T1D.


Subject(s)
Diabetes Mellitus, Type 1 , Disease Progression , Islets of Langerhans , Machine Learning , Single-Cell Analysis , Transcriptome , Humans , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Single-Cell Analysis/methods , Islets of Langerhans/metabolism , Islets of Langerhans/immunology , Transcriptome/genetics , Autoantibodies/immunology , Gene Expression Profiling/methods , Male , Female , Insulin-Secreting Cells/metabolism , Adult
6.
Cells ; 13(5)2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38474380

ABSTRACT

Islet transplantation is a therapeutic option to replace ß-cell mass lost during type 1 or type 3c diabetes. Innate immune responses, particularly the instant blood-mediated inflammatory reaction and activation of monocytes, play a major role in the loss of transplanted islet tissue. In this study, we aimed to investigate the inhibition of toll-like receptor 4 (TLR4) on innate inflammatory responses. We first demonstrate a significant loss of graft function shortly after transplant through the assessment of miR-375 and miR-200c in plasma as biomarkers. Using in vitro models, we investigate how targeting TLR4 mitigates islet damage and immune cell activation during the peritransplant period. The results of this study support the application of TAK-242 as a therapeutic agent to reduce inflammatory and innate immune responses to islets immediately following transplantation into the hepatic portal vein. Therefore, TLR4 may serve as a target to improve islet transplant outcomes in the future.


Subject(s)
Immunity, Innate , Islets of Langerhans Transplantation , Islets of Langerhans , MicroRNAs , Sulfonamides , Toll-Like Receptor 4 , Immunity, Innate/drug effects , Islets of Langerhans Transplantation/methods , Sulfonamides/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Islets of Langerhans/drug effects , Islets of Langerhans/immunology , Humans
7.
Eur J Nutr ; 63(4): 1329-1338, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38413484

ABSTRACT

PURPOSE: The aim was to study the association between dietary intake of B vitamins in childhood and the risk of islet autoimmunity (IA) and progression to type 1 diabetes (T1D) by the age of 10 years. METHODS: We followed 8500 T1D-susceptible children born in the U.S., Finland, Sweden, and Germany in 2004 -2010 from the Environmental Determinants of Diabetes in the Young (TEDDY) study, which is a prospective observational birth cohort. Dietary intake of seven B vitamins was calculated from foods and dietary supplements based on 24-h recall at 3 months and 3-day food records collected regularly from 6 months to 10 years of age. Cox proportional hazard models were adjusted for energy, HLA-genotype, first-degree relative with T1D, sex, and country. RESULTS: A total of 778 (9.2) children developed at least one autoantibody (any IA), and 335 (3.9%) developed multiple autoantibodies. 280 (3.3%) children had IAA and 319 (3.8%) GADA as the first autoantibody. 344 (44%) children with IA progressed to T1D. We observed that higher intake of niacin was associated with a decreased risk of developing multiple autoantibodies (HR 0.95; 95% CI 0.92, 0.98) per 1 mg/1000 kcal in niacin intake. Higher intake of pyridoxine (HR 0.66; 95% CI 0.46, 0.96) and vitamin B12 (HR 0.87; 95% CI 0.77, 0.97) was associated with a decreased risk of IAA-first autoimmunity. Higher intake of riboflavin (HR 1.38; 95% CI 1.05, 1.80) was associated with an increased risk of GADA-first autoimmunity. There were no associations between any of the B vitamins and the outcomes "any IA" and progression from IA to T1D.  CONCLUSION: In this multinational, prospective birth cohort of children with genetic susceptibility to T1D, we observed some direct and inverse associations between different B vitamins and risk of IA.


Subject(s)
Autoantibodies , Autoimmunity , Diabetes Mellitus, Type 1 , Islets of Langerhans , Vitamin B Complex , Humans , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/epidemiology , Male , Female , Vitamin B Complex/administration & dosage , Prospective Studies , Child , Child, Preschool , Infant , Islets of Langerhans/immunology , Autoantibodies/blood , Risk Factors , Diet/methods , Diet/statistics & numerical data , Proportional Hazards Models , United States/epidemiology , Finland/epidemiology , Sweden/epidemiology , Germany/epidemiology , Dietary Supplements , Birth Cohort , Disease Progression
8.
Transplantation ; 108(5): 1115-1126, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38192025

ABSTRACT

BACKGROUND: The utilization of islet-like cells derived from pluripotent stem cells may resolve the scarcity of islet transplantation donors. The subcutaneous space is a promising transplantation site because of its capacity for graft observation and removal, thereby ensuring safety. To guarantee subcutaneous islet transplantation, physicians should ensure ample blood supply. Numerous methodologies, including prevascularization, have been investigated to augment blood flow, but the optimal approach remains undetermined. METHODS: From C57BL/6 mice, 500 syngeneic islets were transplanted into the prevascularized subcutaneous site of recipient mice by implanting agarose rods with basic fibroblast growth factor at 1 and 2 wk. Before transplantation, the blood glucose levels, cell infiltration, and cytokine levels at the transplant site were evaluated. Furthermore, we examined the impact of the extracellular matrix capsule on graft function and the inflammatory response. RESULTS: Compared with the 1-wk group, the 2-wk group exhibited improved glycemic control, indicating that longer prevascularization enhanced transplant success. Flow cytometry analysis detected immune cells, such as neutrophils and macrophages, in the extracellular matrix capsules, whereas cytometric bead array analysis indicated the release of inflammatory and proinflammatory cytokines. Treatment with antitumor necrosis factor and anti-interleukin-6R antibodies in the 1-wk group improved graft survival, similar to the 2-wk group. CONCLUSIONS: In early prevascularization before subcutaneous transplantation, neutrophil and macrophage accumulation prevented early engraftment owing to inflammatory cytokine production.


Subject(s)
Blood Glucose , Cytokines , Graft Survival , Islets of Langerhans Transplantation , Mice, Inbred C57BL , Islets of Langerhans Transplantation/methods , Islets of Langerhans Transplantation/immunology , Animals , Blood Glucose/metabolism , Cytokines/metabolism , Mice , Male , Time Factors , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/surgery , Subcutaneous Tissue/blood supply , Subcutaneous Tissue/immunology , Extracellular Matrix/metabolism , Islets of Langerhans/immunology , Islets of Langerhans/blood supply , Neovascularization, Physiologic
9.
JAMA ; 330(12): 1151-1160, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37682551

ABSTRACT

Importance: The incidence of diabetes in childhood has increased during the COVID-19 pandemic. Elucidating whether SARS-CoV-2 infection is associated with islet autoimmunity, which precedes type 1 diabetes onset, is relevant to disease etiology and future childhood diabetes trends. Objective: To determine whether there is a temporal relationship between SARS-CoV-2 infection and the development of islet autoimmunity in early childhood. Design, Setting, and Participants: Between February 2018 and March 2021, the Primary Oral Insulin Trial, a European multicenter study, enrolled 1050 infants (517 girls) aged 4 to 7 months with a more than 10% genetically defined risk of type 1 diabetes. Children were followed up through September 2022. Exposure: SARS-CoV-2 infection identified by SARS-CoV-2 antibody development in follow-up visits conducted at 2- to 6-month intervals until age 2 years from April 2018 through June 2022. Main Outcomes and Measures: The development of multiple (≥2) islet autoantibodies in follow-up in consecutive samples or single islet antibodies and type 1 diabetes. Antibody incidence rates and risk of developing islet autoantibodies were analyzed. Results: Consent was obtained for 885 (441 girls) children who were included in follow-up antibody measurements from age 6 months. SARS-CoV-2 antibodies developed in 170 children at a median age of 18 months (range, 6-25 months). Islet autoantibodies developed in 60 children. Six of these children tested positive for islet autoantibodies at the same time as they tested positive for SARS-CoV-2 antibodies and 6 at the visit after having tested positive for SARS-CoV-2 antibodies. The sex-, age-, and country-adjusted hazard ratio for developing islet autoantibodies when the children tested positive for SARS-CoV-2 antibodies was 3.5 (95% CI, 1.6-7.7; P = .002). The incidence rate of islet autoantibodies was 3.5 (95% CI, 2.2-5.1) per 100 person-years in children without SARS-CoV-2 antibodies and 7.8 (95% CI, 5.3-19.0) per 100 person-years in children with SARS-CoV-2 antibodies (P = .02). Islet autoantibody risk in children with SARS-CoV-2 antibodies was associated with younger age (<18 months) of SARS-CoV-2 antibody development (HR, 5.3; 95% CI, 1.5-18.3; P = .009). Conclusion and relevance: In young children with high genetic risk of type 1 diabetes, SARS-CoV-2 infection was temporally associated with the development of islet autoantibodies.


Subject(s)
COVID-19 , Diabetes Mellitus, Type 1 , Islets of Langerhans , Child, Preschool , Female , Humans , Infant , Antibodies, Viral/immunology , Autoantibodies/immunology , Autoimmunity/immunology , COVID-19/complications , COVID-19/immunology , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Pandemics , SARS-CoV-2 , Islets of Langerhans/immunology , Male , Genetic Predisposition to Disease
10.
ACS Biomater Sci Eng ; 9(6): 3522-3534, 2023 06 12.
Article in English | MEDLINE | ID: mdl-37233985

ABSTRACT

Type 1 diabetes (T1D) is a life-threatening condition for which islet transplantation offers a way to extend longevity and vastly improve quality of life, but the degree and duration of success can vary greatly due to the patient's protective immunity against foreign material. The field is in need of cellular engineering modalities to promote a localized, tolerogenic environment to protect transplanted islet tissue. Artificial antigen-presenting cells (aAPCs) can be designed exogenously to mimic immune cells, such as dendritic cells, and administered to patients, allowing greater control over T cell differentiation. As regulatory T cell (Treg) modulation can reduce the activity of cytotoxic T-effector populations, this strategy can be used to promote immune acceptance of both biomaterials and cellular transplants, such as islets. A new class of poly(lactic-co-glycolic acid) (PLGA) and PLGA/PBAE-blend aAPCs containing transforming growth factor beta and conjugated with anti-CD3 and anti-CD28 antibodies, called tolerogenic aAPCs (TolAPCs), are specifically designed to generate a tolerogenic response by inducing Tregs. We characterized TolAPCs' physical and chemical properties via advanced particle imaging and sizing modalities and investigated their impact on the local and systemic immune system across BALB/c and C57BL/6 mouse strains as well as healthy male and female mice via histologic, gene expression, and immunofluorescence staining methods. Strain-specific differences were observed, whereas sex made no difference in the TolAPC response. TolAPCs stimulated the expansion of FOXP3+ Tregs and provided islet cell protection, maintaining improved glucose-stimulated insulin secretion in vitro when co-cultured with cytotoxic CD8+ T cells. We also explored the ability of this TolAPC platform to promote tolerance in a streptozotocin-induced murine T1D C57BL/6 mouse model. We achieved partial islet protection over the first few days following co-injection with PLGA/PBAE TolAPCs; however, grafts failed soon thereafter. Analysis of the local injection site demonstrated that other immune cell types, including APCs and cytotoxic natural killer cells, increased in the islet injection site. While we aimed to promote a localized tolerogenic microenvironment in vivo using biodegradable TolAPCs to induce Tregs and extend islet transplant durability, further TolAPC improvements will be required to both elongate efficacy and control additional immune cell responders.


Subject(s)
Islets of Langerhans , T-Lymphocytes, Regulatory , Islets of Langerhans/immunology , Islets of Langerhans/surgery , Pancreas Transplantation , T-Lymphocytes, Regulatory/immunology , Male , Animals , Mice , Female , Diabetes Mellitus, Type 1/immunology , Immunologic Factors/chemistry , Immunologic Factors/therapeutic use , Particle Size
11.
Front Immunol ; 13: 814203, 2022.
Article in English | MEDLINE | ID: mdl-35145521

ABSTRACT

T cells and B cells have been identified in human and murine islets, but the phenotype and role of islet lymphocytes is unknown. Resident immune populations set the stage for responses to inflammation in the islets during homeostasis and diabetes. Thus, we sought to identify the phenotype and effector function of islet lymphocytes to better understand their role in normal islets and in islets under metabolic stress. Lymphocytes were located in the islet parenchyma, and were comprised of a mix of naïve, activated, and memory T cell and B cell subsets, with an enrichment for regulatory B cell subsets. Use of a Nur77 reporter indicated that CD8 T cells and B cells both received local antigen stimulus, indicating that they responded to antigens present in the islets. Analysis of effector function showed that islet T cells and B cells produced the regulatory cytokine IL-10. The regulatory phenotype of islet T cells and B cells and their response to local antigenic stimuli remained stable under conditions of metabolic stress in the diet induced obesity (DIO) model. T cells present in human islets retained a similar activated and memory phenotype in non-diabetic and T2D donors. Under steady-state conditions, islet T cells and B cells have a regulatory phenotype, and thus may play a protective role in maintaining tissue homeostasis.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Homeostasis/physiology , Islets of Langerhans/immunology , Stress, Physiological/physiology , T-Lymphocytes/immunology , Animals , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 2/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Obesity/immunology , Phenotype
12.
PLoS One ; 17(2): e0261103, 2022.
Article in English | MEDLINE | ID: mdl-35196314

ABSTRACT

A variety of islet autoantibodies (AAbs) can predict and possibly dictate eventual type 1 diabetes (T1D) diagnosis. Upwards of 75% of those with T1D are positive for AAbs against glutamic acid decarboxylase (GAD65 or GAD), a producer of gamma-aminobutyric acid (GABA) in human pancreatic beta cells. Interestingly, bacterial populations within the human gut also express GAD and produce GABA. Evidence suggests that dysbiosis of the microbiome may correlate with T1D pathogenesis and physiology. Therefore, autoimmune linkages between the gut microbiome and islets susceptible to autoimmune attack need to be further elucidated. Utilizing in silico analyses, we show that 25 GAD sequences from human gut bacterial sources show sequence and motif similarities to human beta cell GAD65. Our motif analyses determined that most gut GAD sequences contain the pyroxical dependent decarboxylase (PDD) domain of human GAD65, which is important for its enzymatic activity. Additionally, we showed overlap with known human GAD65 T cell receptor epitopes, which may implicate the immune destruction of beta cells. Thus, we propose a physiological hypothesis in which changes in the gut microbiome in those with T1D result in a release of bacterial GAD, thus causing miseducation of the host immune system. Due to the notable similarities we found between human and bacterial GAD, these deputized immune cells may then target human beta cells leading to the development of T1D.


Subject(s)
Autoantibodies/immunology , Bacteria/enzymology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/microbiology , Gastrointestinal Microbiome/immunology , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/immunology , Animals , Antigen-Presenting Cells/immunology , Computer Simulation , Diabetes Mellitus, Type 1/enzymology , Epitopes, T-Lymphocyte/immunology , Genes, Bacterial , Humans , Islets of Langerhans/enzymology , Islets of Langerhans/immunology , Mice , Pan troglodytes/microbiology , Phylogeny , Protein Domains , Sequence Alignment/methods , gamma-Aminobutyric Acid/metabolism
13.
J Pediatr Endocrinol Metab ; 35(3): 393-397, 2022 Mar 28.
Article in English | MEDLINE | ID: mdl-35026884

ABSTRACT

OBJECTIVES: It has been hypothesized that SARS-CoV-2 may play a role in the development of different forms of diabetes mellitus (DM). The Canary Islands have the highest incidence of type 1 DM (T1DM) reported in Spain (30-35/100,000 children under 14 years/year). In 2020-2021 we observed the highest incidence so far on the island of Gran Canaria, as a result of which we decided to evaluate the possible role of COVID-19 in the increased number of onsets. METHODS: We examined the presence of IgG antibodies against SARS-CoV-2 in children with new onset T1DM between October 2020 and August 2021. We compared recent T1DM incidence with that of the previous 10 years. RESULTS: Forty-two patients were diagnosed with T1DM (48.1/100,000 patients/year), representing a nonsignificant 25.7% increase from the expected incidence. Of the 33 patients who consented to the study, 32 presented negative IgG values, with only one patient reflecting undiagnosed past infection. Forty-four percent of patients presented with ketoacidosis at onset, which was similar to previous years. CONCLUSIONS: We conclude that there is no direct relationship between the increased incidence of T1DM and SARS-CoV-2 in the region. The COVID-19 pandemic did not result in an increased severity of T1DM presentation.


Subject(s)
Antibodies, Viral/blood , COVID-19/epidemiology , Diabetes Mellitus, Type 1/epidemiology , Diabetes Mellitus, Type 1/virology , SARS-CoV-2/immunology , Adolescent , Autoantibodies/blood , Child , Child, Preschool , Diabetes Mellitus, Type 1/immunology , Diabetic Ketoacidosis/epidemiology , Humans , Immunoglobulin G/blood , Infant , Infant, Newborn , Islets of Langerhans/immunology , Spain/epidemiology
14.
Diabetes ; 71(3): 566-577, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35007320

ABSTRACT

Type 1 diabetes in children is heralded by a preclinical phase defined by circulating autoantibodies to pancreatic islet antigens. How islet autoimmunity is initiated and then progresses to clinical diabetes remains poorly understood. Only one study has reported gene expression in specific immune cells of children at risk associated with progression to islet autoimmunity. We analyzed gene expression with RNA sequencing in CD4+ and CD8+ T cells, natural killer (NK) cells, and B cells, and chromatin accessibility by assay for transposase-accessible chromatin sequencing (ATAC-seq) in CD4+ T cells, in five genetically at risk children with islet autoantibodies who progressed to diabetes over a median of 3 years ("progressors") compared with five children matched for sex, age, and HLA-DR who had not progressed ("nonprogressors"). In progressors, differentially expressed genes (DEGs) were largely confined to CD4+ T cells and enriched for cytotoxicity-related genes/pathways. Several top-ranked DEGs were validated in a semi-independent cohort of 13 progressors and 11 nonprogressors. Flow cytometry confirmed that progression was associated with expansion of CD4+ cells with a cytotoxic phenotype. By ATAC-seq, progression was associated with reconfiguration of regulatory chromatin regions in CD4+ cells, some linked to differentially expressed cytotoxicity-related genes. Our findings suggest that cytotoxic CD4+ T cells play a role in promoting progression to type 1 diabetes.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Chromatin/chemistry , Cytotoxicity, Immunologic/genetics , Diabetes Mellitus, Type 1/immunology , Disease Progression , Gene Expression Regulation , Adolescent , Autoimmunity/genetics , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/ultrastructure , CD8-Positive T-Lymphocytes/metabolism , Child , Child, Preschool , Diabetes Mellitus, Type 1/genetics , Genetic Predisposition to Disease , Humans , Islets of Langerhans/immunology , Killer Cells, Natural/metabolism , Sequence Analysis, RNA
15.
Diabetes ; 71(3): 483-496, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35007324

ABSTRACT

The induction of antigen (Ag)-specific tolerance and replacement of islet ß-cells are major ongoing goals for the treatment of type 1 diabetes (T1D). Our group previously showed that a hybrid insulin peptide (2.5HIP) is a critical autoantigen for diabetogenic CD4+ T cells in the NOD mouse model. In this study, we investigated whether induction of Ag-specific tolerance using 2.5HIP-coupled tolerogenic nanoparticles (NPs) could protect diabetic NOD mice from disease recurrence upon syngeneic islet transplantation. Islet graft survival was significantly prolonged in mice treated with 2.5HIP NPs, but not NPs containing the insulin B chain peptide 9-23. Protection in 2.5HIP NP-treated mice was attributed both to the simultaneous induction of anergy in 2.5HIP-specific effector T cells and the expansion of Foxp3+ regulatory T cells specific for the same Ag. Notably, our results indicate that effector function of graft-infiltrating CD4+ and CD8+ T cells specific for other ß-cell epitopes was significantly impaired, suggesting a novel mechanism of therapeutically induced linked suppression. This work establishes that tolerance induction with an HIP can delay recurrent autoimmunity in NOD mice, which could inform the development of an Ag-specific therapy for T1D.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Graft Survival/drug effects , Insulin/administration & dosage , Islets of Langerhans Transplantation/methods , Peptide Fragments/administration & dosage , Animals , Autoantigens/immunology , Autoimmunity/immunology , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/prevention & control , Female , Islets of Langerhans/immunology , Mice , Mice, Inbred NOD , Nanoparticles/administration & dosage , Recurrence
16.
Life Sci ; 288: 120184, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34838848

ABSTRACT

AIMS: Rosmarinic acid (RA) is a polyphenol that occurs in plants of the Lamiaceae family. Phenethyl ester of RA (PERA), a novel RA derivative, has been developed and evaluated in vivo in an animal model of type 1 diabetes (T1D). METHODS: T1D was induced in male C57BL/6 mice using multiple low doses of streptozotocin (STZ) administered intraperitoneally for 5 consecutive days. Intraperitoneal administration of PERA (2.5 mg/kg bw) began from the first STZ injection and continued for 20 days. KEY FINDINGS: PERA-treated mice exhibited lower incidence of T1D (monitored up to 38 days from the disease induction), and fluorescent histochemical analysis showed that their pancreatic islets expressed more insulin. PERA treatment significantly down-regulated the proportions of CD11b+ and CD11c+ myeloid cells in the immune cell infiltrates in the pancreatic islets early during T1D pathogenesis (on day 9 after T1D induction), while on day 15, PERA significantly reduced the proportions of CD11c+, CD8+, Th1 and Th17 cells. Simultaneously, it was found that the cells from the pancreatic infiltrates of PERA-treated mice produced significantly less reactive oxygen species than cells from the control group. SIGNIFICANCE: These findings suggest that PERA efficiently prevented T1D development in mice. Interestingly, PERA attenuated the inflammatory process in the islets through temporally specific interference with the innate and adaptive immune response and therefore shows great promise for further clinical evaluation as a novel T1D therapeutic.


Subject(s)
Autoimmunity , Cinnamates/pharmacology , Depsides/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/prevention & control , Esters/chemistry , Islets of Langerhans/drug effects , Animals , Cinnamates/chemistry , Depsides/chemistry , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL , Phenylethyl Alcohol/chemistry , Rosmarinic Acid
17.
Am J Physiol Endocrinol Metab ; 322(2): E109-E117, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34927459

ABSTRACT

Islet transplantation is a treatment for selected adults with type 1 diabetes and severe hypoglycemia. Islets from two or more donor pancreases, a scarce resource, are usually required to impact glycemic control, but the treatment falls short of a cure. Islets are avascular when transplanted into the hypoxic liver environment and subjected to inflammatory insults, immune attack, and toxicity from systemic immunosuppression. The Collaborative Islet Transplant Registry, with outcome data on over 1,000 islet transplant recipients, has demonstrated that larger islet numbers transplanted and older age of recipients are associated with better outcomes. Induction with T-cell depleting agents and the TNF-α inhibitor etanercept and maintenance systemic immunosuppression with mTOR inhibitors in combination with calcineurin inhibitors also appear advantageous, but concerns remain over immunosuppressive toxicity. We discuss strategies and therapeutics that address specific challenges of islet transplantation, many of which are at the preclinical stage of development. On the horizon are adjuvant cell therapies with mesenchymal stromal cells and regulatory T cells that have been used in preclinical models and in humans in other contexts; such a strategy may enable reductions in immunosuppression in the early peri-transplant period when the islets are vulnerable to apoptosis. Human embryonic stem cell-derived islets are in early-phase clinical trials and hold the promise of an inexhaustible supply of insulin-producing cells; effective encapsulation of such cells or, silencing of the human leukocyte antigen (HLA) complex would eliminate the need for immunosuppression, enabling this therapy to be used in all those with type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Hypoglycemia/therapy , Islets of Langerhans Transplantation/methods , Islets of Langerhans/blood supply , Islets of Langerhans/immunology , Registries , Adult , Humans , Immunosuppression Therapy/adverse effects , Islets of Langerhans/drug effects , MTOR Inhibitors/adverse effects , Middle Aged , Transplantation, Homologous/methods , Treatment Outcome
18.
Front Immunol ; 12: 777788, 2021.
Article in English | MEDLINE | ID: mdl-34868047

ABSTRACT

T cell receptors (TCRs) are unique markers that define antigen specificity for a given T cell. With the evolution of sequencing and computational analysis technologies, TCRs are now prime candidates for the development of next-generation non-cell based T cell biomarkers, which provide a surrogate measure to assess the presence of antigen-specific T cells. Type 1 diabetes (T1D), the immune-mediated form of diabetes, is a prototypical organ specific autoimmune disease in which T cells play a pivotal role in targeting pancreatic insulin-producing beta cells. While the disease is now predictable by measuring autoantibodies in the peripheral blood directed to beta cell proteins, there is an urgent need to develop T cell markers that recapitulate T cell activity in the pancreas and can be a measure of disease activity. This review focuses on the potential and challenges of developing TCR biomarkers for T1D. We summarize current knowledge about TCR repertoires and clonotypes specific for T1D and discuss challenges that are unique for autoimmune diabetes. Ultimately, the integration of large TCR datasets produced from individuals with and without T1D along with computational 'big data' analysis will facilitate the development of TCRs as potentially powerful biomarkers in the development of T1D.


Subject(s)
Biomarkers , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/metabolism , Disease Susceptibility , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Alleles , Animals , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/therapy , Epitopes/chemistry , Epitopes/immunology , Epitopes/metabolism , Genetic Predisposition to Disease , Genetic Variation , Histocompatibility Antigens/genetics , Histocompatibility Antigens/immunology , Humans , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Peptides/immunology , Peptides/metabolism , Protein Binding , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology
19.
J Leukoc Biol ; 110(6): 1023-1031, 2021 12.
Article in English | MEDLINE | ID: mdl-34643294

ABSTRACT

The 78-kDa glucose-regulated protein (GRP78) has extracellular, anti-inflammatory properties that can aid resolving inflammation. It has been established previously that GRP78 induced myeloid CD11c+ cell differentiation into distinct tolerogenic cells. This tolerance induction makes GRP78 a potential therapeutic agent for transplanted allogeneic grafts and autoimmune diseases, such as type 1 diabetes. In this research, it is revealed that rmGRP78-treated NOD mice bone marrow-derived CD11c+ cells (GRP78-DCs) highly expressed B7-H4 but down-regulated CD86 and CD40, and retained a tolerogenic signature even after stimulation by LPS. In the assessment of in vivo therapeutic efficacy after the adoptive transfer of GRP78-DCs into NOD mice, fluorescent imaging analyses revealed that the transfer specifically homed in inflamed pancreases, promoting ß-cell survival and alleviating insulitis in NOD mice. The adoptive transfer of GRP78-DCs also helped reduce Th1, Th17, and CTL, suppressing inflammatory cytokine production in vivo. The findings suggest that adoptive GRP78-DC transfer is critical to resolving inflammation in NOD mice and may have relevance in a clinical setting.


Subject(s)
Dendritic Cells/transplantation , Diabetes Mellitus, Type 1/immunology , Endoplasmic Reticulum Chaperone BiP , Immune Tolerance/immunology , Islets of Langerhans , Adoptive Transfer , Animals , Cell Differentiation/drug effects , Cell Differentiation/immunology , Dendritic Cells/immunology , Endoplasmic Reticulum Chaperone BiP/immunology , Endoplasmic Reticulum Chaperone BiP/metabolism , Endoplasmic Reticulum Chaperone BiP/pharmacology , Female , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Mice , Mice, Inbred NOD , Pancreatitis/immunology
20.
Diabet Med ; 38(12): e14717, 2021 12.
Article in English | MEDLINE | ID: mdl-34655243

ABSTRACT

Recent success in clinical trials to delay the onset of type 1 diabetes has heralded a new era of type 1 diabetes research focused on the most accurate methods to predict risk and progression rate in the general population. Risk prediction for type 1 diabetes has been ongoing since the 1970s and 1980s when human leucocyte antigen (HLA) variants and islet autoantibodies associated with type 1 diabetes were first described. Development of prediction methodologies has relied on well-characterised cohorts and samples. The Bart's Oxford (BOX) study of type 1 diabetes has been recruiting children with type 1 diabetes and their first (and second)-degree relatives since 1985. In this review, we use the timeline of the study to review the accompanying basic science developments which have facilitated improved prediction by genetic (HLA analysis through to genetic risk scores) and biochemical strategies (islet cell autoantibodies through to improved individual tests for antibodies to insulin, glutamate decarboxylase, the tyrosine phosphatase IA-2, zinc transporter 8 and tetraspanin 7). The type 1 diabetes community are poised to move forward using the best predictive markers to predict and delay the onset of type 1 diabetes.


Subject(s)
Autoantibodies/immunology , Autoimmunity , Biomedical Research/trends , Diabetes Mellitus, Type 1/immunology , HLA Antigens/immunology , Islets of Langerhans/immunology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...