Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Appl Opt ; 59(12): 3582-3588, 2020 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-32400477

RESUMO

Freeform illumination is one of the necessary techniques in 28 nm technology nodes and beyond. The micromirror array (MMA) has been widely used in lithography freeform illumination systems due to its programmability and high free degree. The MMA allocation algorithm is the key to generate the target freeform illumination source. Its computational speed and precision affect the generation speed and precision of the target illumination source as well as the process window size of the generated illumination pupil directly. In this paper, an MMA allocation method based on deconvolution is proposed. The target freeform illumination source can be obtained directly with the deconvolution and quantization processes. Without the iterative optimization process, the computational speed of the proposed method is much faster than that of the traditional method. The numerical simulation results show that the difference between the target source and the MMA source generated using the proposed method is less than 0.2%. Compared with the process window loss of the target source, the process window loss of the MMA source generated by the proposed deconvolution method is less than 0.5%. Compared with the traditional allocation method, the runtime of the proposed method is less than 0.05 s and has improved by 1463 times.

2.
Diabetes ; 68(5): 988-1001, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30833470

RESUMO

Type 1 diabetes studies consistently generate data showing islet ß-cell dysfunction and T cell-mediated anti-ß-cell-specific autoimmunity. To explore the pathogenesis, we interrogated the ß-cell transcriptomes from donors with and without type 1 diabetes using both bulk-sorted and single ß-cells. Consistent with immunohistological studies, ß-cells from donors with type 1 diabetes displayed increased Class I transcripts and associated mRNA species. These ß-cells also expressed mRNA for Class II and Class II antigen presentation pathway components, but lacked the macrophage marker CD68. Immunohistological study of three independent cohorts of donors with recent-onset type 1 diabetes showed Class II protein and its transcriptional regulator Class II MHC trans-activator protein expressed by a subset of insulin+CD68- ß-cells, specifically found in islets with lymphocytic infiltrates. ß-Cell surface expression of HLA Class II was detected on a portion of CD45-insulin+ ß-cells from donors with type 1 diabetes by immunofluorescence and flow cytometry. Our data demonstrate that pancreatic ß-cells from donors with type 1 diabetes express Class II molecules on selected cells with other key genes in those pathways and inflammation-associated genes. ß-Cell expression of Class II molecules suggests that ß-cells may interact directly with islet-infiltrating CD4+ T cells and may play an immunopathogenic role.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/metabolismo , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/metabolismo , Apresentação de Antígeno/imunologia , Autoimunidade/fisiologia , Linfócitos T CD4-Positivos/metabolismo , Células Cultivadas , Humanos , Insulina/metabolismo
3.
Opt Express ; 27(4): 4629-4647, 2019 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-30876077

RESUMO

Mueller matrix imaging polarimeter (MMIP) can be used to measure the polarization aberration (PA) of a lithographic projector in the form of the Mueller pupil, while the Jones pupil is required for lithographic imaging simulations, projection lens design and PA evaluation. In this paper, a Jones pupil measurement method of lithographic projection lens is proposed. The measurement device of the method is the same as an MMIP, but a new polarimetric measurement equation is derived to solve the Jones pupil directly from the Kronecker product of the Jones matrix and the measured intensities. Two new polarimeter configurations with the minimum condition number are designed to further improve the accuracy in the presence of error sources. The performance of the method is evaluated by measurement errors of a typical Jones pupil in the presence of error sources. Comparisons between the proposed method and the conventional method, in which the Jones pupil is converted from the Mueller pupil measured by MMIP, are given. The results validate that the measurement accuracy of the Jones pupil is significantly improved without increasing the complexity of existing measurement systems.

4.
J Vis Exp ; (136)2018 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-30010652

RESUMO

Experiments using isolated pancreatic islets are important for diabetes research, but islets are expensive and of limited abundance. Islets contain a mixed cell population in a structured architecture that impacts function, and human islets are widely variable in cell type composition. Current frequently used methods to study cultured islets include molecular studies performed on whole islets, lumping disparate islet cell types together, or microscopy or molecular studies on dispersed islet cells, disrupting islet architecture. For in vivo islet studies, paraffin-embedded pancreas sectioning is a powerful technique to assess cell-specific outcomes in the native pancreatic environment. Studying post-culture islets by paraffin sectioning would offer several advantages: detection of multiple outcomes on the same islets (potentially even the exact-same islets, using serial sections), cell-type-specific measurements, and maintaining native islet cell-cell and cell-substratum interactions both during experimental exposure and for analysis. However, existing techniques for embedding isolated islets post-culture are inefficient, time consuming, prone to loss of material, and generally produce sections with inadequate islet numbers to be useful for quantifying outcomes. Clinical pathology laboratory cell block preparation facilities are inaccessible and impractical for basic research laboratories. We have developed an improved, simplified bench-top method that generates sections with robust yield and distribution of islets. Fixed islets are resuspended in warm histological agarose gel and pipetted into a flat disc on a standard glass slide, such that the islets are distributed in a plane. After standard dehydration and embedding, multiple (10+) 4 - 5 µm sections can be cut from the same islet block. Using this method, histological and immunofluorescent analyses can be performed on mouse, rat, and human islets. This is an effective, inexpensive, time-saving approach to assess cell-type-specific, intact-architecture outcomes from cultured islets.


Assuntos
Técnicas Histológicas/métodos , Ilhotas Pancreáticas/metabolismo , Parafina/metabolismo , Animais , Humanos , Ilhotas Pancreáticas/citologia , Camundongos , Parafina/análise , Ratos
5.
PLoS One ; 12(6): e0178641, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28605395

RESUMO

BACKGROUND/AIMS: Studies of human cadaveric pancreas specimens indicate that pancreas inflammation plays an important role in type 1 diabetes pathogenesis. Due to the inaccessibility of pancreas in living patients, imaging technology to visualize pancreas inflammation is much in need. In this study, we investigated the feasibility of utilizing ultrasound imaging to assess pancreas inflammation longitudinally in living rats during the progression leading to type 1 diabetes onset. METHODS: The virus-inducible BBDR type 1 diabetes rat model was used to systematically investigate pancreas changes that occur prior to and during development of autoimmunity. The nearly 100% diabetes incidence upon virus induction and the highly consistent time course of this rat model make longitudinal imaging examination possible. A combination of histology, immunoblotting, flow cytometry, and ultrasound imaging technology was used to identify stage-specific pancreas changes. RESULTS: Our histology data indicated that exocrine pancreas tissue of the diabetes-induced rats underwent dramatic changes, including blood vessel dilation and increased CD8+ cell infiltration, at a very early stage of disease initiation. Ultrasound imaging data revealed significant acute and persistent pancreas inflammation in the diabetes-induced rats. The pancreas micro-vasculature was significantly dilated one day after diabetes induction, and large blood vessel (superior mesenteric artery in this study) dilation and inflammation occurred several days later, but still prior to any observable autoimmune cell infiltration of the pancreatic islets. CONCLUSIONS: Our data demonstrate that ultrasound imaging technology can detect pancreas inflammation in living rats during the development of type 1 diabetes. Due to ultrasound's established use as a non-invasive diagnostic tool, it may prove useful in a clinical setting for type 1 diabetes risk prediction prior to autoimmunity and to assess the effectiveness of potential therapeutics.


Assuntos
Diabetes Mellitus Tipo 1/diagnóstico por imagem , Diabetes Mellitus Tipo 1/patologia , Pancreatite/diagnóstico por imagem , Pancreatite/patologia , Ultrassonografia , Animais , Apoptose , Resistência Capilar , Caspase 3/metabolismo , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1/etiologia , Modelos Animais de Doenças , Humanos , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Microvasos , Pâncreas/irrigação sanguínea , Pâncreas/metabolismo , Pâncreas/patologia , Pancreatite/complicações , Pancreatite/metabolismo , Prognóstico , Ratos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/patologia , Ultrassonografia/métodos
6.
Curr Opin Endocrinol Diabetes Obes ; 24(2): 92-97, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28099206

RESUMO

PURPOSE OF REVIEW: This report examines recent publications identifying phenotypic and functional heterogeneity among pancreatic ß cells and investigating their potential roles in normal and abnormal islet function. The development of new methods and tools for the study of individual islet cells has produced a surge of interest in this topic. RECENT FINDINGS: Studies of ß cell maturation and pregnancy-induced proliferation have identified changes in serotonin and transcription factors SIX2/3 expression as markers of temporal heterogeneity. Structural and functional heterogeneity in the form of functionally distinct 'hub' and 'follower' ß cells was found in mouse islets. Heterogeneous expression of Fltp (in mouse ß cells) and ST8SIA1 and CD9 (in human ß cells) were associated with distinct functional potential. Several impressive reports describing the transcriptomes of individual ß cells were also published in recent months. Some of these reveal previously unknown ß cell subpopulations. SUMMARY: A wealth of information on functional and phenotypic heterogeneity has been collected recently, including the transcriptomes of individual ß cells and the identities of functionally distinct ß cell subpopulations. Several studies suggest the existence of two broad categories: a more proliferative but less functional and a less proliferative but more functional ß cell type. The identification of functionally distinct subpopulations and their association with type 2 diabetes underlines the potential clinical importance of these investigations.


Assuntos
Diferenciação Celular , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/citologia , Animais , Biomarcadores/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia
7.
Diabetes ; 66(1): 145-157, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27999109

RESUMO

The pathogenesis of human type 1 diabetes, characterized by immune-mediated damage of insulin-producing ß-cells of pancreatic islets, may involve viral infection. Essential components of the innate immune antiviral response, including type I interferon (IFN) and IFN receptor-mediated signaling pathways, are candidates for determining susceptibility to human type 1 diabetes. Numerous aspects of human type 1 diabetes pathogenesis are recapitulated in the LEW.1WR1 rat model. Diabetes can be induced in LEW.1WR1 weanling rats challenged with virus or with the viral mimetic polyinosinic:polycytidylic acid (poly I:C). We hypothesized that disrupting the cognate type I IFN receptor (type I IFN α/ß receptor [IFNAR]) to interrupt IFN signaling would prevent or delay the development of virus-induced diabetes. We generated IFNAR1 subunit-deficient LEW.1WR1 rats using CRISPR-Cas9 (clustered regularly interspaced short palindromic repeats-associated protein 9) genome editing and confirmed functional disruption of the Ifnar1 gene. IFNAR1 deficiency significantly delayed the onset and frequency of diabetes and greatly reduced the intensity of insulitis after poly I:C treatment. The occurrence of Kilham rat virus-induced diabetes was also diminished in IFNAR1-deficient animals. These findings firmly establish that alterations in innate immunity influence the course of autoimmune diabetes and support the use of targeted strategies to limit or prevent the development of type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Animais , Sistemas CRISPR-Cas/genética , Sistemas CRISPR-Cas/fisiologia , Células Cultivadas , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/virologia , Feminino , Imunidade Inata/genética , Imunidade Inata/fisiologia , Interferon Tipo I/metabolismo , Masculino , Parvovirus/genética , Parvovirus/fisiologia , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Receptor de Interferon alfa e beta/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Genome Res ; 26(10): 1397-1410, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27470110

RESUMO

RNA-seq protocols that focus on transcript termini are well suited for applications in which template quantity is limiting. Here we show that, when applied to end-sequencing data, analytical methods designed for global RNA-seq produce computational artifacts. To remedy this, we created the End Sequence Analysis Toolkit (ESAT). As a test, we first compared end-sequencing and bulk RNA-seq using RNA from dendritic cells stimulated with lipopolysaccharide (LPS). As predicted by the telescripting model for transcriptional bursts, ESAT detected an LPS-stimulated shift to shorter 3'-isoforms that was not evident by conventional computational methods. Then, droplet-based microfluidics was used to generate 1000 cDNA libraries, each from an individual pancreatic islet cell. ESAT identified nine distinct cell types, three distinct ß-cell types, and a complex interplay between hormone secretion and vascularization. ESAT, then, offers a much-needed and generally applicable computational pipeline for either bulk or single-cell RNA end-sequencing.


Assuntos
Ilhotas Pancreáticas/citologia , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos , Transcriptoma , Animais , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Biblioteca Gênica , Ilhotas Pancreáticas/metabolismo , Microfluídica/métodos , Ratos , Análise de Sequência de RNA/normas , Análise de Célula Única/normas
9.
FASEB J ; 30(2): 983-93, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26546129

RESUMO

Individuals with schizophrenia and their first-degree relatives have higher rates of type 2 diabetes (T2D) than the general population (18-30 vs. 1.2-6.3%), independent of body mass index and antipsychotic medication, suggesting shared genetic components may contribute to both diseases. The cause of this association remains unknown. Mutations in disrupted in schizophrenia 1 (DISC1) increase the risk of developing psychiatric disorders [logarithm (base 10) of odds = 7.1]. Here, we identified DISC1 as a major player controlling pancreatic ß-cell proliferation and insulin secretion via regulation of glycogen synthase kinase-3ß (GSK3ß). DISC1 expression was enriched in developing mouse and human pancreas and adult ß- and ductal cells. Loss of DISC1 function, through siRNA-mediated depletion or expression of a dominant-negative truncation that models the chromosomal translocation of human DISC1 in schizophrenia, resulted in decreased ß-cell proliferation (3 vs. 1%; P < 0.01), increased apoptosis (0.1 vs. 0.6%; P < 0.01), and glucose intolerance in transgenic mice. Insulin secretion was reduced (0.5 vs. 0.1 ng/ml; P < 0.05), and critical ß-cell transcription factors Pdx1 and Nkx6.1 were significantly decreased. Impaired DISC1 allowed inappropriate activation of GSK3ß in ß cells, and antagonizing GSK3ß (SB216763; IC50 = 34.3 nM) rescued the ß-cell defects. These results uncover an unexpected role for DISC1 in normal ß-cell physiology and suggest that DISC1 dysregulation contributes to T2D independently of its importance for cognition.


Assuntos
Proliferação de Células , Quinase 3 da Glicogênio Sintase/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Regulação da Expressão Gênica , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Humanos , Células Secretoras de Insulina/patologia , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Esquizofrenia/genética , Esquizofrenia/metabolismo , Esquizofrenia/patologia , Translocação Genética
10.
Diabetes Metab Syndr Obes ; 8: 387-98, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26316789

RESUMO

OBJECTIVE: Glucagon-like peptide-1 induces glucose-dependent insulin secretion and, in rodents, increases proliferation and survival of pancreatic beta cells. To investigate the effects on human beta cells, we used immunodeficient mice transplanted with human islets. The goal was to determine whether lixisenatide, a glucagon-like peptide-1 receptor agonist, improves human islet function and survival in vivo. METHODS: Five independent transplant studies were conducted with human islets from five individual donors. Diabetic human islet-engrafted immunodeficient mice were treated with lixisenatide (50, 150, and 500 µg/kg) or vehicle. Islet function was determined by blood glucose, plasma human insulin/C-peptide, and glucose tolerance tests. Grafts were analyzed for total beta- and alpha-cell number, percent proliferation, and levels of apoptosis. RESULTS: Diabetic mice transplanted with marginal human islet mass and treated with lixisenatide were restored to euglycemia more rapidly than vehicle-treated mice. Glucose tolerance tests, human plasma insulin, and glucose-stimulation indices of lixisenatide-treated mice were significantly improved compared to vehicle-treated mice. The percentages of proliferating or apoptotic beta cells at graft recovery were not different between lixisenatide-treated and vehicle-treated mice. Nevertheless, in one experiment we found a significant twofold to threefold increase in human beta-cell numbers in lixisenatide-treated compared to vehicle-treated mice. CONCLUSION: Diabetic human islet-engrafted immunodeficient mice treated with lixisenatide show improved restoration of normoglycemia, human plasma insulin, and glucose tolerance compared to vehicle-treated mice engrafted with the same donor islets. Because the proliferative capacity of human beta cells is limited, improved beta-cell survival coupled with enhanced beta-cell function following lixisenatide treatment may provide the greatest benefit for diabetic patients with reduced functional islet mass.

11.
Stem Cell Reports ; 4(4): 591-604, 2015 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-25843049

RESUMO

Human pluripotent stem cells (hPSCs) represent a renewable source of pancreatic beta cells for both basic research and therapeutic applications. Given this outstanding potential, significant efforts have been made to identify the signaling pathways that regulate pancreatic development in hPSC differentiation cultures. In this study, we demonstrate that the combination of epidermal growth factor (EGF) and nicotinamide signaling induces the generation of NKX6-1(+) progenitors from all hPSC lines tested. Furthermore, we show that the size of the NKX6-1(+) population is regulated by the duration of treatment with retinoic acid, fibroblast growth factor 10 (FGF10), and inhibitors of bone morphogenetic protein (BMP) and hedgehog signaling pathways. When transplanted into NOD scid gamma (NSG) recipients, these progenitors differentiate to give rise to exocrine and endocrine cells, including monohormonal insulin(+) cells. Together, these findings provide an efficient and reproducible strategy for generating highly enriched populations of hPSC-derived beta cell progenitors for studies aimed at further characterizing their developmental potential in vivo and deciphering the pathways that regulate their maturation in vitro.


Assuntos
Diferenciação Celular , Proteínas de Homeodomínio/metabolismo , Pâncreas/citologia , Pâncreas/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Ativinas/metabolismo , Animais , Proteínas de Transporte/metabolismo , Linhagem Celular , Fator de Crescimento Epidérmico/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Humanos , Imunofenotipagem , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Camundongos , Modelos Biológicos , Niacinamida/farmacologia , Organogênese/efeitos dos fármacos , Organogênese/genética , Transdução de Sinais/efeitos dos fármacos
12.
Diabetes Metab Syndr Obes ; 6: 493-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24376359

RESUMO

PURPOSE: Dipeptidyl-peptidase-4 (DPP-4) inhibitors are known to increase insulin secretion and beta cell proliferation in rodents. To investigate the effects on human beta cells in vivo, we utilize immunodeficient mice transplanted with human islets. The study goal was to determine the efficacy of alogliptin, a DPP-4 inhibitor, to enhance human beta cell function and proliferation in an in vivo context using diabetic immunodeficient mice engrafted with human pancreatic islets. METHODS: Streptozotocin-induced diabetic NOD-scid IL2rγ(null) (NSG) mice were transplanted with adult human islets in three separate trials. Transplanted mice were treated daily by gavage with alogliptin (30 mg/kg/day) or vehicle control. Islet graft function was compared using glucose tolerance tests and non-fasting plasma levels of human insulin and C-peptide; beta cell proliferation was determined by bromodeoxyuridine (BrdU) incorporation. RESULTS: Glucose tolerance tests were significantly improved by alogliptin treatment for mice transplanted with islets from two of the three human islet donors. Islet-engrafted mice treated with alogliptin also had significantly higher plasma levels of human insulin and C-peptide compared to vehicle controls. The percentage of insulin+BrdU+ cells in human islet grafts from alogliptin-treated mice was approximately 10-fold more than from vehicle control mice, consistent with a significant increase in human beta cell proliferation. CONCLUSION: Human islet-engrafted immunodeficient mice treated with alogliptin show improved human insulin secretion and beta cell proliferation compared to control mice engrafted with the same donor islets. Immunodeficient mice transplanted with human islets provide a useful model to interrogate potential therapies to improve human islet function and survival in vivo.

13.
Diabetologia ; 56(12): 2638-46, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24121653

RESUMO

AIMS/HYPOTHESIS: We hypothesised that pathological endoplasmic reticulum (ER) stress contributes to beta cell death during development of type 1 diabetes. In this study, we investigated the occurrence of beta cell ER stress and the signalling pathways involved during discrete stages of autoimmune diabetes progression. The virus-inducible BBDR rat model was used to systematically interrogate the three main ER stress signalling pathways (IRE1 [inositol-requiring protein-1], PERK [double-stranded RNA-dependent protein kinase (PKR)-like ER kinase] and ATF6 [activating transcription factor 6]) in pancreatic beta cells during type 1 diabetes development. METHODS: ER stress and apoptotic markers were assessed by immunoblot analyses of isolated pancreatic islets and immunofluorescence staining of pancreas sections from control and virus-induced rats. Various time points were analysed: (1) early stages preceding the development of insulitis and (2) a late stage during onset and progression of insulitis, which precedes overt hyperglycaemia. RESULTS: The IRE1 pathway, including its downstream component X-box-binding protein 1, was specifically activated in pancreatic beta cells of virus-induced rats at early stages preceding the development of insulitis. Furthermore, ER stress-specific pro-apoptotic caspase 12 and effector caspase 3 were also activated at this stage. Activation of PERK and its downstream effector pro-apoptotic CHOP (CCAAT/-enhancer-binding-protein homologous protein), only occurred during late stages of diabetes induction concurrent with insulitis, whereas ATF6 activation in pancreatic beta cells was similar in control and virus-induced rats. CONCLUSIONS/INTERPRETATION: Activation of the IRE1 pathway and ER stress-specific pro-apoptotic caspase 12, before the development of insulitis, are indicative of ER stress-mediated beta cell damage. The early occurrence of pathological ER stress and death in pancreatic beta cells may contribute to the initiation and/or progression of virus-induced autoimmune diabetes.


Assuntos
Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/patologia , Estresse do Retículo Endoplasmático , Células Secretoras de Insulina/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fator 6 Ativador da Transcrição/metabolismo , Animais , Apoptose , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Feminino , Immunoblotting , Células Secretoras de Insulina/patologia , Masculino , Ratos , Transdução de Sinais , Fator de Transcrição CHOP/metabolismo , eIF-2 Quinase/metabolismo
14.
PLoS One ; 8(10): e78050, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24147110

RESUMO

Epidemiologic and clinical evidence suggests that virus infection plays an important role in human type 1 diabetes pathogenesis. We used the virus-inducible BioBreeding Diabetes Resistant (BBDR) rat to investigate the ability of sodium salicylate, a non-steroidal anti-inflammatory drug (NSAID), to modulate development of type 1 diabetes. BBDR rats treated with Kilham rat virus (KRV) and polyinosinic:polycytidylic acid (pIC, a TLR3 agonist) develop diabetes at nearly 100% incidence by ~2 weeks. We found distinct temporal profiles of the proinflammatory serum cytokines, IL-1ß, IL-6, IFN-γ, IL-12, and haptoglobin (an acute phase protein) in KRV+pIC treated rats. Significant elevations of IL-1ß and IL-12, coupled with sustained elevations of haptoglobin, were specific to KRV+pIC and not found in rats co-treated with pIC and H1, a non-diabetogenic virus. Salicylate administered concurrently with KRV+pIC inhibited the elevations in IL-1ß, IL-6, IFN-γ and haptoglobin almost completely, and reduced IL-12 levels significantly. Salicylate prevented diabetes in a dose-dependent manner, and diabetes-free animals had no evidence of insulitis. Our data support an important role for innate immunity in virus-induced type 1 diabetes pathogenesis. The ability of salicylate to prevent diabetes in this robust animal model demonstrates its potential use to prevent or attenuate human autoimmune diabetes.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/virologia , Salicilatos/uso terapêutico , Animais , Diabetes Mellitus Tipo 1/induzido quimicamente , Feminino , Masculino , Parvovirus/patogenicidade , Poli I-C/toxicidade , Ratos , Ratos Sprague-Dawley
15.
Methods Mol Biol ; 933: 31-44, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22893399

RESUMO

The BB rat is an important rodent model of human type 1 diabetes (T1D) and has been used to study mechanisms of diabetes pathogenesis as well as to investigate potential intervention therapies for clinical trials. The Diabetes-Prone BB (BBDP) rat spontaneously develops autoimmune T1D between 50 and 90 days of age. The Diabetes-Resistant BB (BBDR) rat has similar diabetes-susceptible genes as the BBDP, but does not become diabetic in viral antibody-free conditions. However, the BBDR rat can be induced to develop T1D in response to certain treatments such as regulatory T cell (T(reg)) depletion, toll-like receptor ligation, or virus infection. These diabetes-inducible rats develop hyperglycemia under well-controlled circumstances and within a short, predictable time frame (14-21 days), thus facilitating their utility for investigations of specific stages of diabetes development. Therefore, these rat strains are invaluable models for studying autoimmune diabetes and the role of environmental factors in its development, of particular importance due to the influx of studies associating virus infection and human T1D.


Assuntos
Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Modelos Animais de Doenças , Ratos Endogâmicos BB , Animais , Autoimunidade , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Experimental/virologia , Diabetes Mellitus Tipo 1/terapia , Diabetes Mellitus Tipo 1/virologia , Humanos , Hiperglicemia/etiologia , Imunidade Inata , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/patologia , Ratos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Receptores Toll-Like/imunologia , Viroses/complicações , Viroses/imunologia , Vírus/imunologia , Vírus/isolamento & purificação
17.
Autoimmunity ; 44(2): 137-48, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20695765

RESUMO

The adipokine, leptin, regulates blood glucose and the insulin secretory function of beta cells, while also modulating immune cell function. We hypothesized that the dual effects of leptin may prevent or suppress the autoreactive destruction of beta cells in a virally induced rodent model of type 1 diabetes. Nearly 100% of weanling BBDR rats treated with the combination of an innate immune system activator, polyinosinic:polycytidylic acid (pIC), and Kilham rat virus (KRV) become diabetic within a predictable time frame. We utilized this model to test the efficacy of leptin in preventing diabetes onset, remitting new onset disease, and preventing autoimmune recurrence in diabetic rats transplanted with syngeneic islet grafts. High doses of leptin delivered via an adenovirus vector (AdLeptin) or alzet pump prevented diabetes in>90% of rats treated with pIC+KRV. The serum hyperleptinemia generated by this treatment was associated with decreased body weight, decreased non-fasting serum insulin levels, and lack of islet insulitis in leptin-treated rats. In new onset diabetics, hyperleptinemia prevented rapid weight loss and diabetic ketoacidosis, and temporarily restored euglycemia. Leptin treatment also prolonged the survival of syngeneic islets transplanted into diabetic BBDR rats. In diverse therapeutic settings, we found leptin treatment to have significant beneficial effects in modulating virally induced diabetes. These findings merit further evaluation of leptin as a potential adjunct therapeutic agent for treatment of human type 1 diabetes.


Assuntos
Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Tipo 1/tratamento farmacológico , Leptina/uso terapêutico , Infecções por Parvoviridae/imunologia , Parvovirus/imunologia , Animais , Glicemia , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/virologia , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/virologia , Cetoacidose Diabética/prevenção & controle , Humanos , Transplante das Ilhotas Pancreáticas , Leptina/administração & dosagem , Leptina/imunologia , Infecções por Parvoviridae/virologia , Poli I-C/administração & dosagem , Poli I-C/imunologia , Ratos , Ratos Endogâmicos BB , Resultado do Tratamento
18.
Gen Comp Endocrinol ; 170(2): 334-45, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20965191

RESUMO

Zebrafish embryos are emerging as models of glucose metabolism. However, patterns of endogenous glucose levels, and the role of the islet in glucoregulation, are unknown. We measured absolute glucose levels in zebrafish and mouse embryos, and demonstrate similar, dynamic glucose fluctuations in both species. Further, we show that chemical and genetic perturbations elicit mammalian-like glycemic responses in zebrafish embryos. We show that glucose is undetectable in early zebrafish and mouse embryos, but increases in parallel with pancreatic islet formation in both species. In zebrafish, increasing glucose is associated with activation of gluconeogenic phosphoenolpyruvate carboxykinase1 (pck1) transcription. Non-hepatic Pck1 protein is expressed in mouse embryos. We show using RNA in situ hybridization, that zebrafish pck1 mRNA is similarly expressed in multiple cell types prior to hepatogenesis. Further, we demonstrate that the Pck1 inhibitor 3-mercaptopicolinic acid suppresses normal glucose accumulation in early zebrafish embryos. This shows that pre- and extra-hepatic pck1 is functional, and provides glucose locally to rapidly developing tissues. To determine if the primary islet is glucoregulatory in early fish embryos, we injected pdx1-specific morpholinos into transgenic embryos expressing GFP in beta cells. Most morphant islets were hypomorphic, not a genetic, but embryos still exhibited persistent hyperglycemia. We conclude from these data that the early zebrafish islet is functional, and regulates endogenous glucose. In summary, we identify mechanisms of glucoregulation in zebrafish embryos that are conserved with embryonic and adult mammals. These observations justify use of this model in mechanistic studies of human metabolic disease.


Assuntos
Embrião não Mamífero/metabolismo , Glucose/metabolismo , Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Desenvolvimento Embrionário/efeitos dos fármacos , Proteínas de Fluorescência Verde/análise , Hibridização In Situ , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Fosfoenolpiruvato Carboxiquinase (GTP)/genética , Fosfoenolpiruvato Carboxiquinase (GTP)/metabolismo , Fosfoenolpiruvato Carboxiquinase (GTP)/fisiologia , Filogenia , Ácidos Picolínicos/farmacologia , RNA Mensageiro/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento
19.
Exp Biol Med (Maywood) ; 235(11): 1328-37, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20975081

RESUMO

Proteomic profiling of serum is a powerful technique to identify differentially expressed proteins that can serve as biomarkers predictive of disease onset. In this study, we utilized two-dimensional (2D) gel analysis followed by matrix-assisted-laser desorption/ionization time-of-flight mass spectrometry analysis to identify putative serum biomarkers for autoimmune type 1 diabetes (T1D) in biobreeding diabetes resistant (BBDR) rats induced to express the disease. Treatment with toll-like receptor 3 ligand, polyinosinic:polycytidilic acid (pIC), plus infection with Kilham rat virus (KRV), a rat parvovirus, results in nearly 100% of young BBDR rats becoming diabetic within 11-21 d. Sera collected from prediabetic rats at early time points following treatment with pIC + KRV were analyzed by 2D gel electrophoresis and compared with sera from control rats treated with phosphate-buffered saline, pIC alone or pIC + H1, a non-diabetogenic parvovirus. None of the latter three control treatments precipitates T1D. 2D gel analysis revealed that haptoglobin, an acute phase and hemoglobin scavenger protein, was differentially expressed in the sera of rats treated with pIC + KRV relative to control groups. These results were confirmed by Western blot and enzyme-linked immunosorbent assay studies, which further validated haptoglobin levels as being differentially increased in the sera of pIC + KRV-treated rats relative to controls during the first week following infection. Early elevations in serum haptoglobin were also observed in LEW1.WR1 rats that became diabetic following infection with rat cytomegalovirus. The identification and validation of haptoglobin as a putative serum biomarker for autoimmune T1D in rats now affords us the opportunity to test the validity of this protein as a biomarker for human T1D, particularly in those situations where viral infection is believed to precede the onset of disease.


Assuntos
Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/virologia , Haptoglobinas/análise , Animais , Biomarcadores/sangue , Western Blotting , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/diagnóstico , Diabetes Mellitus Experimental/virologia , Diabetes Mellitus Tipo 1/sangue , Eletroforese em Gel Bidimensional , Ensaio de Imunoadsorção Enzimática , Feminino , Masculino , Parvovirus , Poli I-C , Ratos , Ratos Endogâmicos Lew , Ratos Endogâmicos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
20.
Biophys J ; 99(5): 1546-55, 2010 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-20816067

RESUMO

We are investigating the influence of the converter and relay domains on elementary rate constants of the actomyosin cross-bridge cycle. The converter and relay domains vary between Drosophila myosin heavy chain isoforms due to alternative mRNA splicing. Previously, we found that separate insertions of embryonic myosin isoform (EMB) versions of these domains into the indirect flight muscle (IFM) myosin isoform (IFI) both decreased Drosophila IFM power and slowed muscle kinetics. To determine cross-bridge mechanisms behind the changes, we employed sinusoidal analysis while varying phosphate and MgATP concentrations in skinned Drosophila IFM fibers. Based on a six-state cross-bridge model, the EMB converter decreased myosin rate constants associated with actin attachment and work production, k(4), but increased rates related to cross-bridge detachment and work absorption, k(2). In contrast, the EMB relay domain had little influence on kinetics, because only k(4) decreased. The main alteration was mechanical, in that work production amplitude decreased. That both domains decreased k(4) supports the hypothesis that these domains are critical to lever-arm-mediated force generation. Neither domain significantly influenced MgATP affinity. Our modeling suggests the converter domain is responsible for the difference in rate-limiting cross-bridge steps between EMB and IFI myosin--i.e., a myosin isomerization associated with MgADP release for EMB and Pi release for IFI.


Assuntos
Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Voo Animal , Músculo Esquelético/metabolismo , Miosinas/química , Miosinas/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Drosophila melanogaster/fisiologia , Cinética , Modelos Moleculares , Fosfatos/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA