Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Nat Commun ; 15(1): 3744, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702321

RESUMEN

Cellular composition and anatomical organization influence normal and aberrant organ functions. Emerging spatial single-cell proteomic assays such as Image Mass Cytometry (IMC) and Co-Detection by Indexing (CODEX) have facilitated the study of cellular composition and organization by enabling high-throughput measurement of cells and their localization directly in intact tissues. However, annotation of cell types and quantification of their relative localization in tissues remain challenging. To address these unmet needs for atlas-scale datasets like Human Pancreas Analysis Program (HPAP), we develop AnnoSpat (Annotator and Spatial Pattern Finder) that uses neural network and point process algorithms to automatically identify cell types and quantify cell-cell proximity relationships. Our study of data from IMC and CODEX shows the higher performance of AnnoSpat in rapid and accurate annotation of cell types compared to alternative approaches. Moreover, the application of AnnoSpat to type 1 diabetic, non-diabetic autoantibody-positive, and non-diabetic organ donor cohorts recapitulates known islet pathobiology and shows differential dynamics of pancreatic polypeptide (PP) cell abundance and CD8+ T cells infiltration in islets during type 1 diabetes progression.


Asunto(s)
Algoritmos , Diabetes Mellitus Tipo 1 , Páncreas , Proteómica , Humanos , Proteómica/métodos , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/metabolismo , Páncreas/citología , Páncreas/metabolismo , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/citología , Análisis de la Célula Individual/métodos , Redes Neurales de la Computación , Linfocitos T CD8-positivos/metabolismo , Citometría de Imagen/métodos
2.
Nat Cell Biol ; 26(3): 421-437, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38409327

RESUMEN

Type 1 diabetes (T1D) is characterized by the destruction of pancreatic ß-cells. Several observations have renewed the interest in ß-cell RNA sensors and editors. Here, we report that N6-methyladenosine (m6A) is an adaptive ß-cell safeguard mechanism that controls the amplitude and duration of the antiviral innate immune response at T1D onset. m6A writer methyltransferase 3 (METTL3) levels increase drastically in ß-cells at T1D onset but rapidly decline with disease progression. m6A sequencing revealed the m6A hypermethylation of several key innate immune mediators, including OAS1, OAS2, OAS3 and ADAR1 in human islets and EndoC-ßH1 cells at T1D onset. METTL3 silencing enhanced 2'-5'-oligoadenylate synthetase levels by increasing its mRNA stability. Consistently, in vivo gene therapy to prolong Mettl3 overexpression specifically in ß-cells delayed diabetes progression in the non-obese diabetic mouse model of T1D. Mechanistically, the accumulation of reactive oxygen species blocked upregulation of METTL3 in response to cytokines, while physiological levels of nitric oxide enhanced METTL3 levels and activity. Furthermore, we report that the cysteines in position C276 and C326 in the zinc finger domains of the METTL3 protein are sensitive to S-nitrosylation and are important to the METTL3-mediated regulation of oligoadenylate synthase mRNA stability in human ß-cells. Collectively, we report that m6A regulates the innate immune response at the ß-cell level during the onset of T1D in humans.


Asunto(s)
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Animales , Humanos , Ratones , Adenosina Desaminasa/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Inmunidad Innata , Células Secretoras de Insulina/metabolismo , Metiltransferasas/genética , Metiltransferasas/metabolismo , Oxidación-Reducción
3.
Cell Metab ; 36(1): 48-61.e6, 2024 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-38128529

RESUMEN

A major hypothesis for the etiology of type 1 diabetes (T1D) postulates initiation by viral infection, leading to double-stranded RNA (dsRNA)-mediated interferon response and inflammation; however, a causal virus has not been identified. Here, we use a mouse model, corroborated with human islet data, to demonstrate that endogenous dsRNA in beta cells can lead to a diabetogenic immune response, thus identifying a virus-independent mechanism for T1D initiation. We found that disruption of the RNA editing enzyme adenosine deaminases acting on RNA (ADAR) in beta cells triggers a massive interferon response, islet inflammation, and beta cell failure and destruction, with features bearing striking similarity to early-stage human T1D. Glycolysis via calcium enhances the interferon response, suggesting an actionable vicious cycle of inflammation and increased beta cell workload.


Asunto(s)
Diabetes Mellitus Tipo 1 , Ratones , Animales , Humanos , Edición de ARN , ARN Bicatenario , Interferones/genética , Interferones/metabolismo , Inflamación
4.
Diabetes ; 2023 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-37881846

RESUMEN

The endocrine and exocrine compartments of the pancreas are spatially related but functionally distinct. Multiple diseases affect both compartments, including type 1 diabetes (T1D), pancreatitis, cystic fibrosis, and pancreatic cancer. To better understand how the exocrine pancreas changes with age, obesity, and diabetes, we performed systematic analysis of wellpreserved tissue sections from the pancreatic head, body, and tail of organ donors with T1D (n = 20), type 2 diabetes (T2D, n = 25), and donors with no diabetes (ND, n = 74). Among ND donors, we found that acinar-to-ductal metaplasia (ADM), angiopathy, and pancreatic adiposity increased with age, while ADM and adiposity also increased with BMI. Compared to age- and sex-matched ND organs, T1D pancreata had greater acinar atrophy and angiopathy with fewer intralobular adipocytes. T2D pancreata had greater ADM, angiopathy, and total T lymphocytes, but no difference in adipocyte number, compared to ND organs. While total pancreatic fibrosis was increased in both T1D and T2D, the pattern was different with T1D pancreata having greater periductal and perivascular fibrosis, whereas T2D pancreata had greater lobular and parenchymal fibrosis. Thus, the exocrine pancreas undergoes distinct changes as individuals age or develop T1D or T2D.

5.
bioRxiv ; 2023 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-36824909

RESUMEN

Type 1 Diabetes (T1D) is characterized by autoimmune-mediated destruction of insulin-producing ß-cells. Several observations have renewed interest in the innate immune system as an initiator of the disease process against ß-cells. Here, we show that N 6 -Methyladenosine (m 6 A) is an adaptive ß-cell safeguard mechanism that accelerates mRNA decay of the 2'-5'-oligoadenylate synthetase (OAS) genes to control the antiviral innate immune response at T1D onset. m 6 A writer methyltransferase 3 (METTL3) levels increase drastically in human and mouse ß-cells at T1D onset but rapidly decline with disease progression. Treatment of human islets and EndoC-ßH1 cells with pro-inflammatory cytokines interleukin-1 ß and interferon α mimicked the METTL3 upregulation seen at T1D onset. Furthermore, m 6 A-sequencing revealed the m 6 A hypermethylation of several key innate immune mediators including OAS1, OAS2, and OAS3 in human islets and EndoC-ßH1 cells challenged with cytokines. METTL3 silencing in human pseudoislets or EndoC-ßH1 cells enhanced OAS levels by increasing its mRNA stability upon cytokine challenge. Consistently, in vivo gene therapy, to prolong Mettl3 overexpression specifically in ß-cells, delayed diabetes progression in the non-obese diabetic (NOD) mouse model of T1D by limiting the upregulation of Oas pointing to potential therapeutic relevance. Mechanistically, the accumulation of reactive oxygen species blocked METTL3 upregulation in response to cytokines, while physiological levels of nitric oxide promoted its expression in human islets. Furthermore, for the first time to our knowledge, we show that the cysteines in position C276 and C326 in the zinc finger domain of the METTL3 protein are sensitive to S-nitrosylation (SNO) and are significant for the METTL3 mediated regulation of OAS mRNA stability in human ß-cells in response to cytokines. Collectively, we report that m 6 A regulates human and mouse ß-cells to control the innate immune response during the onset of T1D and propose targeting METTL3 to prevent ß-cell death in T1D.

6.
bioRxiv ; 2023 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-36712052

RESUMEN

Cellular composition and anatomical organization influence normal and aberrant organ functions. Emerging spatial single-cell proteomic assays such as Image Mass Cytometry (IMC) and Co-Detection by Indexing (CODEX) have facilitated the study of cellular composition and organization by enabling high-throughput measurement of cells and their localization directly in intact tissues. However, annotation of cell types and quantification of their relative localization in tissues remain challenging. To address these unmet needs, we developed AnnoSpat (Annotator and Spatial Pattern Finder) that uses neural network and point process algorithms to automatically identify cell types and quantify cell-cell proximity relationships. Our study of data from IMC and CODEX show the superior performance of AnnoSpat in rapid and accurate annotation of cell types compared to alternative approaches. Moreover, the application of AnnoSpat to type 1 diabetic, non-diabetic autoantibody-positive, and non-diabetic organ donor cohorts recapitulated known islet pathobiology and showed differential dynamics of pancreatic polypeptide (PP) cell abundance and CD8+ T cells infiltration in islets during type 1 diabetes progression.

8.
Cell Metab ; 33(11): 2103-2105, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34731653

RESUMEN

Transplantation of insulin-producing cells is an emerging treatment for type 1 diabetes. A recent report in Cell Stem Cell (Aghazadeh et al., 2021) outlines a new approach that accelerates the engraftment and improves the survival and function of such cell transplants by mixing adipose tissue-derived ready-made microvessels with human pancreatic progenitor cells or cadaveric islets prior to transplantation.


Asunto(s)
Diabetes Mellitus Tipo 1 , Insulinas , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Diabetes Mellitus Tipo 1/terapia , Humanos , Microvasos
9.
Nat Rev Endocrinol ; 17(7): 389-399, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33875857

RESUMEN

Immune checkpoint inhibitors (ICIs) are monoclonal antibodies that target two key signalling pathways related to T cell activation and exhaustion, by binding to and inhibiting cytotoxic T lymphocyte antigen 4 (CTLA4) or PD1 and its ligand PDL1. ICIs, such as nivolumab, pembrolizumab and ipilimumab, are approved for the treatment of numerous and diverse cancer types, in various combination regimens, and are now an established cornerstone of cancer therapeutics. Toxicities induced by ICIs are autoimmune in nature and are referred to as immune-related adverse events (irAEs); these events can affect any organ system in an unpredictable fashion. Importantly, irAEs can manifest as endocrinopathies involving the thyroid (hypothyroidism or thyrotoxicosis), pituitary (hypophysitis), adrenal glands (adrenal insufficiency) and pancreas (diabetes mellitus). These events are a frequent source of acute and persistent morbidity in patients treated with ICIs and can even be fatal. Over the past few years, there has been a growing understanding of the underlying pathogenesis of irAEs that has led to the development of more effective management strategies. Herein, we review the current understanding of the pathobiology, clinical manifestations and treatment approaches to endocrine toxicities arising from ICIs.


Asunto(s)
Enfermedades del Sistema Endocrino/inducido químicamente , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Antineoplásicos Inmunológicos/efectos adversos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/epidemiología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/inmunología , Enfermedades del Sistema Endocrino/diagnóstico , Enfermedades del Sistema Endocrino/epidemiología , Enfermedades del Sistema Endocrino/terapia , Humanos , Sistema Inmunológico/efectos de los fármacos , Sistema Inmunológico/fisiología , Inmunoterapia/efectos adversos , Neoplasias/tratamiento farmacológico , Neoplasias/epidemiología , Neoplasias/inmunología
10.
Diabetes ; 69(3): 342-354, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31836690

RESUMEN

Human but not mouse islets transplanted into immunodeficient NSG mice effectively accumulate lipid droplets (LDs). Because chronic lipid exposure is associated with islet ß-cell dysfunction, we investigated LD accumulation in the intact human and mouse pancreas over a range of ages and states of diabetes. Very few LDs were found in normal human juvenile pancreatic acinar and islet cells, with numbers subsequently increasing throughout adulthood. While accumulation appeared evenly distributed in postjuvenile acinar and islet cells in donors without diabetes, LDs were enriched in islet α- and ß-cells from donors with type 2 diabetes (T2D). LDs were also found in the islet ß-like cells produced from human embryonic cell-derived ß-cell clusters. In contrast, LD accumulation was nearly undetectable in the adult rodent pancreas, even in hyperglycemic and hyperlipidemic models or 1.5-year-old mice. Taken together, there appear to be significant differences in pancreas islet cell lipid handling between species, and the human juvenile and adult cell populations. Moreover, our results suggest that LD enrichment could be impactful to T2D islet cell function.


Asunto(s)
Diabetes Mellitus Tipo 2/patología , Células Secretoras de Glucagón/patología , Células Secretoras de Insulina/patología , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/patología , Gotas Lipídicas/patología , Células Acinares/patología , Células Acinares/ultraestructura , Adolescente , Adulto , Factores de Edad , Anciano , Animales , Niño , Preescolar , Diabetes Mellitus Experimental/patología , Células Madre Embrionarias , Femenino , Células Secretoras de Glucagón/ultraestructura , Humanos , Lactante , Células Secretoras de Insulina/ultraestructura , Islotes Pancreáticos/citología , Islotes Pancreáticos/ultraestructura , Gotas Lipídicas/ultraestructura , Masculino , Ratones , Microscopía Electrónica , Microscopía Fluorescente , Persona de Mediana Edad , Ratas , Donantes de Tejidos , Adulto Joven
12.
Cell Metab ; 30(5): 976-986.e3, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31495689

RESUMEN

The importance of pancreatic versus intestinal-derived GLP-1 for glucose homeostasis is controversial. We detected active GLP-1 in the mouse and human pancreas, albeit at extremely low levels relative to glucagon. Accordingly, to elucidate the metabolic importance of intestinal proglucagon-derived peptides (PGDPs), we generated mice with reduction of Gcg expression within the distal (GcgDistalGut-/-) or entire (GcgGut-/-) gut. Substantial reduction of gut Gcg expression markedly reduced circulating levels of GLP-1, and impaired glucose homeostasis, associated with increased levels of GIP, and accelerated gastric emptying. GcgDistalGut-/- mice similarly exhibited lower circulating GLP-1 and impaired oral glucose tolerance. Nevertheless, plasma levels of insulin remained normal following glucose administration in the absence of gut-derived GLP-1. Collectively, our findings identify the essential importance of gut-derived PGDPs for maintaining levels of circulating GLP-1, control of gastric emptying, and glucose homeostasis.


Asunto(s)
Microbioma Gastrointestinal , Animales , Glucosa , Homeostasis , Humanos , Insulina , Ratones , Péptidos , Proglucagón
13.
Diabetes ; 68(7): 1394-1402, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31127054

RESUMEN

Type 1 diabetes risk can reliably be predicted by markers of autoimmunity, but approaches to prevent or modify the underlying disease process are needed. We posit this void fundamentally results from a limited understanding of immune-islet cell interactions within the pancreas and relevant immune organs, contributions of ß-cells to their own demise, and epigenetic predispositions affecting both immune and islet cells. Because biopsy of the human pancreas and pancreatic lymph nodes carries risk and the pancreas begins to autodigest soon after death, detailed cellular and molecular phenotyping of the human type 1 diabetes pancreas is lacking, limiting our understanding of the mechanisms of ß-cell loss. To address these challenges, the National Institutes of Health/National Institute of Diabetes and Digestive and Kidney Diseases established the Human Pancreas Analysis Program (HPAP) to procure human type 1 diabetes pancreata for an extensive array of tissue-based, cellular, and epigenetic assays aimed at critical knowledge gaps in our understanding of the local immune attack and loss of ß-cells. In this Methodology Review, we describe how HPAP is performing detailed islet and immune cell phenotyping and creating publicly available data sets with the goals of an improved understanding of type 1 diabetes and the development of more effective treatments to prevent or reverse the disease.


Asunto(s)
Autoinmunidad/inmunología , Diabetes Mellitus Tipo 1/inmunología , Células Secretoras de Insulina/inmunología , Insulina/metabolismo , Páncreas/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , National Institutes of Health (U.S.) , Páncreas/metabolismo , Estados Unidos
14.
Biol Blood Marrow Transplant ; 25(6): 1225-1231, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30738170

RESUMEN

New-onset post-transplant diabetes mellitus (PTDM) occurs frequently after allogeneic hematopoietic cell transplant (HCT). Although calcineurin inhibitors and corticosteroids are assumed to be the cause for hyperglycemia, patients developing PTDM have elevated fasting C-peptide levels before HCT and before immunosuppressive medications. To determine if PTDM results from established insulin resistance present before transplant, we performed oral glucose tolerance tests (OGTTs) and measured whole body, peripheral, and hepatic insulin sensitivity with euglycemic hyperinsulinemic clamps before and 90 days after HLA-identical sibling donor HCT in 20 patients without pretransplant diabetes. HCT recipients were prospectively followed for the development of new-onset PTDM defined as a weekly fasting blood glucose ≥ 126 mg/dL or random blood glucose ≥ 200 mg/dL. During the first 100 days all patients received calcineurin inhibitors, and 11 individuals (55%) were prospectively diagnosed with new-onset PTDM. PTDM diagnosis preceded corticosteroid treatment. During the pretransplant OGTT, elevated fasting (87 mg/dL versus 101 mg/dL; P = .005) but not 2-hour postprandial glucose levels predicted PTDM diagnosis (P = .648). In response to insulin infusion during the euglycemic hyperinsulinemic clamp, patients developing PTDM had lower whole body glucose utilization (P = .047) and decreased peripheral/skeletal muscle uptake (P = .031) before and after transplant, respectively, when compared with non-PTDM patients. Hepatic insulin sensitivity did not differ. Survival was decreased in PTDM patients (2-year estimate, 55% versus 100%; P = .039). Insulin resistance before HCT is a risk factor for PTDM independent of immunosuppression. Fasting pretransplant glucose levels identified PTDM susceptibility, and peripheral insulin resistance could be targeted for prevention and treatment of PTDM after HCT.


Asunto(s)
Diabetes Mellitus/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Acondicionamiento Pretrasplante/efectos adversos , Adulto , Femenino , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Resistencia a la Insulina , Masculino , Persona de Mediana Edad , Acondicionamiento Pretrasplante/métodos , Trasplante Homólogo
15.
Cell Metab ; 29(3): 745-754.e4, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30449685

RESUMEN

Identification of cell-surface markers specific to human pancreatic ß cells would allow in vivo analysis and imaging. Here we introduce a biomarker, ectonucleoside triphosphate diphosphohydrolase-3 (NTPDase3), that is expressed on the cell surface of essentially all adult human ß cells, including those from individuals with type 1 or type 2 diabetes. NTPDase3 is expressed dynamically during postnatal human pancreas development, appearing first in acinar cells at birth, but several months later its expression declines in acinar cells while concurrently emerging in islet ß cells. Given its specificity and membrane localization, we utilized an NTPDase3 antibody for purification of live human ß cells as confirmed by transcriptional profiling, and, in addition, for in vivo imaging of transplanted human ß cells. Thus, NTPDase3 is a cell-surface biomarker of adult human ß cells, and the antibody directed to this protein should be a useful new reagent for ß cell sorting, in vivo imaging, and targeting.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Páncreas/metabolismo , Adulto , Animales , Biomarcadores/metabolismo , Células Cultivadas , Humanos , Células Secretoras de Insulina/patología , Islotes Pancreáticos/patología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos NOD , Páncreas/patología , Adulto Joven
16.
Diabetes ; 67(11): 2305-2318, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30150306

RESUMEN

Type 1 diabetes (T1D) is an autoimmune disease where pancreatic ß-cells are destroyed by islet-infiltrating T cells. Although a role for ß-cell defects has been suspected, ß-cell abnormalities are difficult to demonstrate. We show a ß-cell DNA damage response (DDR), presented by activation of the 53BP1 protein and accumulation of p53, in biopsy and autopsy material from patients with recently diagnosed T1D as well as a rat model of human T1D. The ß-cell DDR is more frequent in islets infiltrated by CD45+ immune cells, suggesting a link to islet inflammation. The ß-cell toxin streptozotocin (STZ) elicits DDR in islets, both in vivo and ex vivo, and causes elevation of the proinflammatory molecules IL-1ß and Cxcl10. ß-Cell-specific inactivation of the master DNA repair gene ataxia telangiectasia mutated (ATM) in STZ-treated mice decreases the expression of proinflammatory cytokines in islets and attenuates the development of hyperglycemia. Together, these data suggest that ß-cell DDR is an early event in T1D, possibly contributing to autoimmunity.


Asunto(s)
Daño del ADN/inmunología , Diabetes Mellitus Tipo 1/inmunología , Inflamación/inmunología , Células Secretoras de Insulina/inmunología , Islotes Pancreáticos/inmunología , Adulto , Animales , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/patología , Femenino , Humanos , Inflamación/patología , Células Secretoras de Insulina/patología , Islotes Pancreáticos/patología , Masculino , Ratones , Persona de Mediana Edad , Adulto Joven
17.
Pancreas ; 47(5): 516-525, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29702529

RESUMEN

A workshop on "The Interface of Pancreatic Cancer with Diabetes, Obesity, and Inflammation: Research Gaps and Opportunities" was held by the National Institute of Diabetes and Digestive and Kidney Diseases on October 12, 2017. The purpose of the workshop was to explore the relationship and possible mechanisms of the increased risk of pancreatic ductal adenocarcinoma (PDAC) related to diabetes, the role of altered intracellular energy metabolism in PDAC, the mechanisms and biomarkers of diabetes caused by PDAC, the mechanisms of the increased risk of PDAC associated with obesity, and the role of inflammatory events and mediators as contributing causes of the development of PDAC. Workshop faculty reviewed the state of the current knowledge in these areas and made recommendations for future research efforts. Further knowledge is needed to elucidate the basic mechanisms contributing to the role of hyperinsulinemia, hyperglycemia, adipokines, and acute and chronic inflammatory events on the development of PDAC.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Diabetes Mellitus/patología , Inflamación/patología , Obesidad/patología , Neoplasias Pancreáticas/patología , Investigación Biomédica/métodos , Investigación Biomédica/tendencias , Carcinoma Ductal Pancreático/epidemiología , Carcinoma Ductal Pancreático/metabolismo , Comorbilidad , Diabetes Mellitus/epidemiología , Diabetes Mellitus/metabolismo , Metabolismo Energético , Humanos , Inflamación/epidemiología , Inflamación/metabolismo , National Institute of Diabetes and Digestive and Kidney Diseases (U.S.) , Obesidad/epidemiología , Obesidad/metabolismo , Neoplasias Pancreáticas/epidemiología , Neoplasias Pancreáticas/metabolismo , Factores de Riesgo , Estados Unidos/epidemiología
18.
Diabetes ; 67(7): 1356-1368, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29654212

RESUMEN

In spite of tolerance mechanisms, some individuals develop T-cell-mediated autoimmunity. Posttranslational modifications that increase the affinity of epitope presentation and/or recognition represent one means through which self-tolerance mechanisms can be circumvented. We investigated T-cell recognition of peptides that correspond to modified ß-cell antigens in subjects with type 1 diabetes. Modified peptides elicited enhanced proliferation by autoreactive T-cell clones. Endoplasmic reticulum (ER) stress in insulinoma cells increased cytosolic calcium and the activity of tissue transglutaminase 2 (tTG2). Furthermore, stressed human islets and insulinomas elicited effector responses from T cells specific for modified peptides, suggesting that ER stress-derived tTG2 activity generated deamidated neoepitopes that autoreactive T cells recognized. Patients with type 1 diabetes had large numbers of T cells specific for these epitopes in their peripheral blood. T cells with these specificities were also isolated from the pancreatic draining lymph nodes of cadaveric donors with established diabetes. Together, these results suggest that self-antigens are enzymatically modified in ß-cells during ER stress, giving rise to modified epitopes that could serve to initiate autoimmunity or to further broaden the antigenic repertoire, activating potentially pathogenic CD4+ T cells that may not be effectively eliminated by negative selection.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Estrés del Retículo Endoplásmico/fisiología , Epítopos de Linfocito T/metabolismo , Células Secretoras de Insulina/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Presentación de Antígeno , Autoantígenos/inmunología , Autoinmunidad/inmunología , Estudios de Casos y Controles , Células Cultivadas , Diabetes Mellitus Tipo 1/metabolismo , Activación Enzimática , Epítopos de Linfocito T/inmunología , Proteínas de Unión al GTP/metabolismo , Humanos , Insectos , Células Secretoras de Insulina/inmunología , Proteína Glutamina Gamma Glutamiltransferasa 2 , Procesamiento Proteico-Postraduccional/fisiología , Arginina Deiminasa Proteína-Tipo 2 , Desiminasas de la Arginina Proteica/metabolismo , Transglutaminasas/metabolismo
19.
JCI Insight ; 3(8)2018 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-29669939

RESUMEN

Cystic fibrosis-related (CF-related) diabetes (CFRD) is an increasingly common and devastating comorbidity of CF, affecting approximately 35% of adults with CF. However, the underlying causes of CFRD are unclear. Here, we examined cystic fibrosis transmembrane conductance regulator (CFTR) islet expression and whether the CFTR participates in islet endocrine cell function using murine models of ß cell CFTR deletion and normal and CF human pancreas and islets. Specific deletion of CFTR from murine ß cells did not affect ß cell function. In human islets, CFTR mRNA was minimally expressed, and CFTR protein and electrical activity were not detected. Isolated CF/CFRD islets demonstrated appropriate insulin and glucagon secretion, with few changes in key islet-regulatory transcripts. Furthermore, approximately 65% of ß cell area was lost in CF donors, compounded by pancreatic remodeling and immune infiltration of the islet. These results indicate that CFRD is caused by ß cell loss and intraislet inflammation in the setting of a complex pleiotropic disease and not by intrinsic islet dysfunction from CFTR mutation.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/etiología , Complicaciones de la Diabetes/genética , Diabetes Mellitus/genética , Islotes Pancreáticos/metabolismo , Adulto , Animales , Fibrosis Quística/genética , Fibrosis Quística/patología , Fibrosis Quística/veterinaria , Complicaciones de la Diabetes/veterinaria , Diabetes Mellitus/epidemiología , Diabetes Mellitus/veterinaria , Femenino , Eliminación de Gen , Glucagón/metabolismo , Humanos , Inflamación/complicaciones , Inflamación/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Masculino , Ratones , Mutación
20.
PLoS One ; 13(1): e0190993, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29324894

RESUMEN

BACKGROUND: Obesity is known to be a major risk factor for diabetes, but the magnitude of risk and variation between blacks and whites are less well documented in populations heavily affected by obesity. Herein we assess rates and risks of incident diabetes in a diverse southern population where obesity is common. METHODS: A total of 24,000 black and 14,064 white adults aged 40-79 in the Southern Community Cohort Study with no self-reported diabetes at study enrollment during 2002-2009 was followed for up to 10 (median 4.5) years. Incidence rates, odds ratios (OR) and accompanying 95% confidence intervals (CI) for medication-treated incident diabetes were determined according to body mass index (BMI) and other characteristics, including tobacco and alcohol consumption, healthy eating and physical activity indices, and socioeconomic status (SES). RESULTS: Risk of incident diabetes rose monotonically with increasing BMI, but the trends differed between blacks and whites (pinteraction < .0001). Adjusted ORs (CIs) for diabetes among those with BMI≥40 vs 20-25 kg/m2 were 11.9 (8.4-16.8) for whites and 4.0 (3.3-4.8) for blacks. Diabetes incidence was more than twice as high among blacks than whites of normal BMI, but the racial difference became attenuated as BMI rose, with estimated 5-year probabilities of developing diabetes approaching 20% for both blacks and whites with BMI≥40 kg/m2. Diabetes risk was also associated with low SES, significantly (pinteraction≤.02) more so for whites, current cigarette smoking, and lower healthy eating and physical activity indices, although high BMI remained the predominant risk factor among both blacks and whites. From baseline prevalence and 20-year projections of the incidence trends, we estimate that the large majority of surviving cohort participants with BMI≥40 kg/m2 will be diagnosed with diabetes. CONCLUSIONS: Even using conservative criteria to ascertain diabetes incidence (i.e., requiring diabetes medication use and ignoring undiagnosed cases), rates of obesity-associated diabetes were exceptionally high in this low-income adult population. The findings indicate that effective strategies to halt the rising prevalence of obesity are needed to avoid substantial increases in diabetes in coming years.


Asunto(s)
Negro o Afroamericano , Diabetes Mellitus/epidemiología , Obesidad/epidemiología , Pobreza , Población Blanca , Adulto , Anciano , Diabetes Mellitus/etnología , Femenino , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Obesidad/complicaciones , Obesidad/etnología , Estados Unidos/epidemiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA