Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 12(1): 9745, 2022 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-35697702

RESUMO

PD-L1 (22C3) checkpoint inhibitor therapy represents a mainstay of modern cancer immunotherapy for non-small cell lung cancer (NSCLC). In vitro diagnostic (IVD) PD-L1 antibody staining is widely used to predict clinical intervention efficacy. However, pathologist interpretation of this assay is cumbersome and variable, resulting in poor positive predictive value concerning patient therapy response. To address this, we developed a digital assay (DA) termed Tissue Insight (TI) 22C3 NSCLC, for the quantification of PD-L1 in NSCLC tissues, including digital recognition of macrophages and lymphocytes. We completed clinical validation of this digital image analysis solution in 66 NSCLC patient samples, followed by concordance studies (comparison of PD-L1 manual and digital scores) in an additional 99 patient samples. We then combined this DA with three distinct immune cell recognition algorithms for detecting tissue macrophages, alveolar macrophages, and lymphocytes to aid in sample interpretation. Our PD-L1 (22C3) DA was successfully validated and had a scoring agreement (digital to manual) higher than the inter-pathologist scoring. Furthermore, the number of algorithm-identified immune cells showed significant correlation when compared with those identified by immunohistochemistry in serial sections stained by double immunofluorescence. Here, we demonstrated that TI 22C3 NSCLC DA yields comparable results to pathologist interpretation while eliminating the intra- and inter-pathologist variability associated with manual scoring while providing characterization of the immune microenvironment, which can aid in clinical treatment decisions.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Algoritmos , Antígeno B7-H1 , Biomarcadores Tumorais/análise , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Microambiente Tumoral
2.
Sci Rep ; 11(1): 9920, 2021 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-33972574

RESUMO

Dickkopf-1 (DKK1) is a secreted modulator of Wnt signaling that is frequently overexpressed in tumors and associated with poor clinical outcomes. DKN-01 is a humanized monoclonal therapeutic antibody that binds DKK1 with high affinity and has demonstrated clinical activity in gastric/gastroesophageal junction (G/GEJ) patients with elevated tumoral expression of DKK1. Here we report on the validation of a DKK1 RNAscope chromogenic in situ hybridization assay to assess DKK1 expression in G/GEJ tumor tissue. To reduce pathologist time, potential pathologist variability from manual scoring and support pathologist decision making, a digital image analysis algorithm that identifies tumor cells and quantifies the DKK1 signal was developed. Following CLIA guidelines the DKK1 RNAscope chromogenic in situ hybridization assay and digital image analysis algorithm were successfully validated for sensitivity, specificity, accuracy, and precision. The DKK1 RNAscope assay in conjunction with the digital image analysis solution is acceptable for prospective screening of G/GEJ adenocarcinoma patients. The work described here will further advance the companion diagnostic development of our DKK1 RNAscope assay and could generally be used as a guide for the validation of RNAscope assays with digital image quantification.


Assuntos
Adenocarcinoma/diagnóstico , Biomarcadores Tumorais/análise , Neoplasias Esofágicas/diagnóstico , Processamento de Imagem Assistida por Computador/métodos , Peptídeos e Proteínas de Sinalização Intercelular/análise , Neoplasias Gástricas/diagnóstico , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/patologia , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Ensaios Clínicos Fase II como Assunto , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Junção Esofagogástrica/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Hibridização In Situ/métodos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Pessoa de Meia-Idade , Estudos Multicêntricos como Assunto , Estudos Prospectivos , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Estudos Retrospectivos , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Via de Sinalização Wnt/efeitos dos fármacos , Via de Sinalização Wnt/genética
3.
Diabetes ; 68(9): 1819-1829, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31167877

RESUMO

We identified autoantibodies (AAb) reacting with a variant IA-2 molecule (IA-2var) that has three amino acid substitutions (Cys27, Gly608, and Pro671) within the full-length molecule. We examined IA-2var AAb in first-degree relatives of type 1 diabetes (T1D) probands from the TrialNet Pathway to Prevention Study. The presence of IA-2var-specific AAb in relatives was associated with accelerated progression to T1D in those positive for AAb to GAD65 and/or insulin but negative in the standard test for IA-2 AAb. Furthermore, relatives with single islet AAb (by traditional assays) and carrying both IA-2var AAb and the high-risk HLA-DRB1*04-DQB1*03:02 haplotype progress rapidly to onset of T1D. Molecular modeling of IA-2var predicts that the genomic variation that alters the three amino acids induces changes in the three-dimensional structure of the molecule, which may lead to epitope unmasking in the IA-2 extracellular domain. Our observations suggest that the presence of AAb to IA-2var would identify high-risk subjects who would benefit from participation in prevention trials who have one islet antibody by traditional testing and otherwise would be misclassified as "low risk" relatives.


Assuntos
Autoanticorpos , Diabetes Mellitus Tipo 1/diagnóstico , Cadeias HLA-DRB1/genética , Proteínas Tirosina Fosfatases Classe 8 Semelhantes a Receptores/imunologia , Adolescente , Adulto , Criança , Pré-Escolar , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Feminino , Haplótipos , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Adulto Jovem
4.
JCI Insight ; 3(23)2018 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-30518692

RESUMO

We describe a protective effect on autoimmune diabetes and reduced destructive insulitis in NOD.scid recipients following splenocyte injections from diabetic NOD donors and sorted CD19+ cells compared with NOD.scid recipients receiving splenocytes alone. This protective effect was age specific (only CD19+ cells from young NOD donors exerted this effect; P < 0.001). We found that the CD19+IgM+ cell is the primary subpopulation of B cells that delayed transfer of diabetes mediated by diabetogenic T cells from NOD mice (P = 0.002). Removal of IgM+ cells from the CD19+ pool did not result in protection. Notably, protection conferred by CD19+IgM+ cotransfers were not dependent on the presence of Tregs, as their depletion did not affect their ability to delay onset of diabetes. Blockade of IL-10 with neutralizing antibodies at the time of CD19+ cell cotransfers also abrogated the therapeutic effect, suggesting that IL-10 secretion was an important component of protection. These results were strengthened by ex vivo incubation of CD19+ cells with IL-5, resulting in enhanced proliferation and IL-10 production and equivalently delayed diabetes progression (P = 0.0005). The potential to expand CD19+IgM+ cells, especially in response to IL-5 stimulation or by pharmacologic agents, may be a new therapeutic option for type 1 diabetes.


Assuntos
Antígenos CD19/uso terapêutico , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/terapia , Imunoglobulina M/uso terapêutico , Animais , Anticorpos Neutralizantes , Linfócitos B , Proliferação de Células , Feminino , Antígenos de Histocompatibilidade Classe I , Interleucina-10/metabolismo , Interleucina-5/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID
5.
Arch Pathol Lab Med ; 141(9): 1267-1275, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28557614

RESUMO

CONTEXT: - Novel therapeutics often target complex cellular mechanisms. Increasingly, quantitative methods like digital tissue image analysis (tIA) are required to evaluate correspondingly complex biomarkers to elucidate subtle phenotypes that can inform treatment decisions with these targeted therapies. These tIA systems need a gold standard, or reference method, to establish analytical validity. Conventional, subjective histopathologic scores assigned by an experienced pathologist are the gold standard in anatomic pathology and are an attractive reference method. The pathologist's score can establish the ground truth to assess a tIA solution's analytical performance. The paradox of this validation strategy, however, is that tIA is often used to assist pathologists to score complex biomarkers because it is more objective and reproducible than manual evaluation alone by overcoming known biases in a human's visual evaluation of tissue, and because it can generate endpoints that cannot be generated by a human observer. OBJECTIVE: - To discuss common visual and cognitive traps known in traditional pathology-based scoring paradigms that may impact characterization of tIA-assisted scoring accuracy, sensitivity, and specificity. DATA SOURCES: - This manuscript reviews the current literature from the past decades available for traditional subjective pathology scoring paradigms and known cognitive and visual traps relevant to these scoring paradigms. CONCLUSIONS: - Awareness of the gold standard paradox is necessary when using traditional pathologist scores to analytically validate a tIA tool because image analysis is used specifically to overcome known sources of bias in visual assessment of tissue sections.


Assuntos
Biomarcadores/análise , Interpretação de Imagem Assistida por Computador/métodos , Imuno-Histoquímica/métodos , Patologia Clínica/métodos , Humanos
6.
Diabetes Care ; 40(4): 561-568, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28174261

RESUMO

OBJECTIVE: The characterization of diverse subtypes of diabetes is a dynamic field of clinical research and an active area of discussion. The objective of this study was to identify new antigenic determinants in the neuroendocrine autoantigen IA-2 (ICA512) and assess whether circulating autoantibodies directed to new IA-2 epitopes identify autoimmune diabetes in young and adult populations with diabetes. RESEARCH DESIGN AND METHODS: Clinically diagnosed patients with type 2 diabetes (n = 258; diabetes duration: 0.01-31 years) were evaluated using a new biomarker detecting autoantibodies directed to the extracellular domain of the neuroendocrine autoantigen IA-2 (IA-2ec). The proportion of IA-2ec autoantibodies was also evaluated in newly diagnosed patients with type 1 diabetes (n = 150; diabetes duration: 0.04-0.49 years). In addition, IA-2 (intracellular domain), GAD65, and zinc transporter 8 autoantibodies were assayed. RESULTS: IA-2ec autoantibodies were detected in patients with type 1 diabetes and, surprisingly, in 5% of patients with type 2 diabetes without serologic responses to other IA-2 antigenic epitopes or other islet autoantigens. We also assessed the ability of IA-2ec-derived peptides to elicit CD4+ T-cell responses by stimulating peripheral blood mononuclear cells from patients with type 1 diabetes (n = 18) and HLA-matched healthy subjects (n = 13) with peptides and staining with the peptide/DQ8-specific tetramers, observing disease-associated responses to previously unreported epitopes within IA-2ec. CONCLUSIONS: We developed a new antibody biomarker identifying novel antigenic determinants within the N terminus of IA-2. IA-2ec autoantibodies can be detected in patients with type 1 diabetes and in a subgroup of adult autoimmune patients with type 2 diabetes phenotype negative for conventional islet autoantibody testing. These observations suggest that islet autoimmunity may be more common in clinically diagnosed type 2 diabetes than previously observed.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Epitopos/imunologia , Proteínas Tirosina Fosfatases Classe 8 Semelhantes a Receptores/imunologia , Adolescente , Adulto , Autoanticorpos/imunologia , Autoantígenos/imunologia , Biomarcadores/análise , Criança , Pré-Escolar , Diabetes Mellitus Tipo 2/imunologia , Feminino , Glutamato Descarboxilase/imunologia , Humanos , Leucócitos Mononucleares/imunologia , Masculino , Pessoa de Meia-Idade , Linfócitos T/imunologia , Adulto Jovem
7.
PLoS One ; 9(9): e106537, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25226365

RESUMO

There is a paucity of literature describing metabolic and histological data in adult-onset autoimmune diabetes. This subgroup of diabetes mellitus affects at least 5% of clinically diagnosed type 2 diabetic patients (T2DM) and it is termed Latent Autoimmune Diabetes in Adults (LADA). We evaluated indexes of insulin secretion, metabolic assessment, and pancreatic pathology in clinically diagnosed T2DM patients with and without the presence of humoral islet autoimmunity (Ab). A total of 18 patients with at least 5-year duration of clinically diagnosed T2DM were evaluated in this study. In those subjects we assessed acute insulin responses to arginine, a glucose clamp study, whole-body fat mass and fat-free mass. We have also analyzed the pancreatic pathology of 15 T2DM and 43 control cadaveric donors, using pancreatic tissue obtained from all the T2DM organ donors available from the nPOD network through December 31, 2013. The presence of islet Ab correlated with severely impaired ß-cell function as demonstrated by remarkably low acute insulin response to arginine (AIR) when compared to that of the Ab negative group. Glucose clamp studies indicated that both Ab positive and Ab negative patients exhibited peripheral insulin resistance in a similar fashion. Pathology data from T2DM donors with Ab or the autoimmune diabetes associated DR3/DR4 allelic class II combination showed reduction in beta cell mass as well as presence of autoimmune-associated pattern A pathology in subjects with either islet autoantibodies or the DR3/DR4 genotype. In conclusion, we provide compelling evidence indicating that islet Ab positive long-term T2DM patients exhibit profound impairment of insulin secretion as well as reduced beta cell mass seemingly determined by an immune-mediated injury of pancreatic ß-cells. Deciphering the mechanisms underlying beta cell destruction in this subset of diabetic patients may lead to the development of novel immunologic therapies aimed at halting the disease progression in its early stage.


Assuntos
Autoimunidade , Diabetes Mellitus Tipo 2/imunologia , Diabetes Mellitus Tipo 2/metabolismo , Resistência à Insulina , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/metabolismo , Adulto , Idoso , Alelos , Autoanticorpos/imunologia , Biomarcadores , Glicemia , Composição Corporal , Peptídeo C/metabolismo , Tamanho Celular , Diabetes Mellitus Tipo 2/genética , Feminino , Antígenos HLA-DR/genética , Antígenos HLA-DR/imunologia , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/patologia , Masculino , Pessoa de Meia-Idade , Doadores de Tecidos
8.
Virology ; 468-470: 19-27, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25129435

RESUMO

We used the LEW1.WR1 rat model of Kilham Rat Virus (KRV)-induced type 1 diabetes (T1D) to test the hypothesis that disease mechanisms are linked with beta cell infection and intra-islet immune activation prior to insulitis. KRV induces genes involved in type I and type II interferon pathways in islet cell lines in vitro and in islets from day-5-infected animals in vivo via mechanisms that do not involve insulitis, beta cell apoptosis, or impaired insulin expression. Immunohistochemistry studies indicated that KRV protein is expressed in beta cells 5 days following infection. KRV induces the phosphorylation of Janus Kinase 1/2 (JAK1/2) and signal transducer and activator of transcription 1 (STAT-1) in islet cells via a mechanism that could involve TLR9 and NF-κB pathways. These data demonstrate for the first time that KRV-induced islet destruction is associated with beta cell infection and intra-islet innate immune upregulation early in the disease process.


Assuntos
Diabetes Mellitus Tipo 1/virologia , Ilhotas Pancreáticas/fisiologia , Janus Quinase 1/metabolismo , Janus Quinase 2/metabolismo , Parvovirus/fisiologia , Fator de Transcrição STAT1/metabolismo , Animais , Linhagem Celular , Diabetes Mellitus Tipo 1/patologia , Inflamação/metabolismo , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/virologia , Janus Quinase 1/genética , Janus Quinase 2/genética , Ratos , Ratos Endogâmicos , Fator de Transcrição STAT1/genética , Transdução de Sinais , Organismos Livres de Patógenos Específicos
9.
Pediatr Diabetes ; 15(1): 1-9, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24325575

RESUMO

nPOD actively promotes a multidisciplinary and unbiased approach toward a better understanding of T1D and identify novel therapeutic targets, through its focus on the study of human samples. Unique to this effort is the coordination of collaborative efforts and real-time data sharing. Studies supported by nPOD are providing direct evidence that human T1D isa complex and heterogeneous disease, in which a multitude of pathogenic factors may be operational and may contribute to the onset of the disease. Importantly, the concept that beta cell destruction is almost completed and that the autoimmune process is almost extinguished soon after diagnosis is being challenged. nPOD investigators are exploring the hypothesis that beta cell dysfunction may also be a significant cause of hyperglycemia, at least around the time of diagnosis, and are uncovering novel molecules and pathways that are linked to the pathogenesis and etiology of human T1D. The validation of therapeutic targets is also a key component of this effort, with recent and future findings providing new strategic direction for clinical trials.


Assuntos
Diabetes Mellitus Tipo 1/fisiopatologia , Doadores de Tecidos , Adulto , Idoso , Autoanticorpos/fisiologia , Comportamento Cooperativo , Diabetes Mellitus Tipo 1/virologia , Feminino , Humanos , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/fisiologia , Masculino , Pessoa de Meia-Idade , Pâncreas/imunologia , Pâncreas/patologia , Transplante de Pâncreas , Regeneração , Bancos de Tecidos , Adulto Jovem
10.
J Diabetes Complications ; 27(4): 307-15, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23558108

RESUMO

AIM: Metabolic syndrome affects a large proportion of the population and increases cardiovascular disease risk. Because metabolic syndrome often co-exists clinically with atherosclerosis, it is difficult to distinguish the respective contributions of the components to vascular abnormalities. Accordingly, we utilized a porcine dietary model of metabolic syndrome without atherosclerosis to investigate early abnormalities of vascular function and signaling. METHODS: Thirty-two Yucatan micropigs were fed either a high-fat, high-simple-sugar, high-calorie (HFHS) or standard chow diet (STD) for 6 months. Neither diet contained added cholesterol. Blood pressure and flow-mediated vasodilatation were assessed at baseline and 6 months. Aortas were harvested at 6 months to assess histology, insulin signaling, and endothelial nitric oxide (eNOS) phosphorylation. RESULTS: HFHS pigs developed characteristics of metabolic syndrome including obesity, dyslipidemia, and insulin resistance, but without histologic evidence of atherosclerosis. Although arterial intima-media thickness did not differ between groups, vascular dysfunction in HFHS was manifest by increased blood pressure and impaired flow-mediated vasodilation of the femoral artery. Compared with STD, aortas from HFHS exhibited increased p85α expression and Ser307 IRS-1 phosphorylation, and blunted insulin-stimulated IRS-1-associated phosphatidylinositol (PI) 3-kinase activity. In the absence of insulin stimulation, aortic Akt Ser473-phosphorylation was greater in HFHS than in STD. With insulin stimulation, Akt phosphorylation increased in STD, but not HFHS. Insulin-induced Ser1177-phosphorylation of eNOS was decreased in HFHS, compared with STD. CONCLUSIONS: Pigs with metabolic syndrome develop early vascular dysfunction and aortic insulin signaling abnormalities, and could be a useful model for early human vascular abnormalities in this condition.


Assuntos
Resistência à Insulina/fisiologia , Síndrome Metabólica/complicações , Obesidade/complicações , Obesidade/metabolismo , Porco Miniatura , Animais , Artérias/metabolismo , Dieta , Modelos Animais de Doenças , Feminino , Proteínas Substratos do Receptor de Insulina/metabolismo , Masculino , Síndrome Metabólica/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Obesidade/etiologia , Fosforilação , Suínos , Vasodilatação
11.
Diabetes Metab Res Rev ; 28(7): 608-17, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22585677

RESUMO

BACKGROUND: The Network for Pancreatic Organ Donors with Diabetes (nPOD) was established to recover and characterize pancreata and related organs from cadaveric organ donors with various risk levels for type 1 diabetes (T1D). These biospecimens are available to investigators for collaborative studies aimed at addressing questions related to T1D natural history and pathogenesis. RESEARCH DESIGN AND METHODS: Organ donors included T1D patients (new onset to long term), non-diabetic autoantibody-positive subjects, non-diabetic controls and individuals with disorders relevant to ß-cell function. Pancreas recovery and transport met transplant-grade criteria. Additional samples recovered included serum, whole blood, spleen and pancreatic and non-pancreatic lymph nodes. Biospecimens were processed for cryopreserved cells, fixed paraffin and fresh frozen blocks and snap frozen samples. T1D autoantibodies, C-peptide levels and high-resolution HLA genotyping for risk alleles were also determined. RESULTS: Over 160 donors have been enrolled (ages of 1 day to >90 years). Standard operating procedures were established along with a quality management system. Donor demographics, laboratory assays and histopathological characterizations were shared through an open online informatics system. Biospecimens were distributed to more than 60 investigators. CONCLUSIONS: The nPOD programme provides access to high quality biospecimens without cost to investigators. Collaborations and open data sharing are emphasized to maximize research potential of each donor. On the basis of initial successes, the nPOD programme is expanding to recover additional organs relevant to T1D pathogenesis and complications from European countries (PanFin network).


Assuntos
Diabetes Mellitus Tipo 1/fisiopatologia , Pâncreas/fisiopatologia , Bancos de Tecidos , Pesquisa Biomédica , Cadáver , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Florida , Humanos , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Pâncreas/imunologia , Pâncreas/patologia , Bancos de Tecidos/organização & administração
12.
Adv Exp Med Biol ; 771: 310-8, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23393687

RESUMO

Diabetes mellitus has been traditionally classified as Type 1 and Type 2 on the basis of several criteria that generally reflect either insulin deficiency or functional defects in insulin secretion. In this chapter, we propose a new classification diabetes based on age of onset, body mass index and biomarkers such as islet autoantibodies and DR HLA alleles. In the second part of this chapter, we briefly discuss some novel hypotheses on the possibility of beta-cell regeneration in diabetes in relation to the islet pathology of the disease.


Assuntos
Diabetes Mellitus Tipo 1/classificação , Diabetes Mellitus Tipo 2/classificação , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/fisiologia , Regeneração/fisiologia , Amiloide/metabolismo , Amiloidose/patologia , Amiloidose/fisiopatologia , Animais , Biomarcadores/metabolismo , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/fisiopatologia , Diabetes Mellitus Tipo 2/patologia , Diabetes Mellitus Tipo 2/fisiopatologia , Humanos
13.
Semin Immunopathol ; 33(1): 23-7, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21188381

RESUMO

The issue of beta cell regeneration in human pancreas is probably one of the most controversial aspects of type 1 diabetes research. In this review, we will first describe the known mechanisms underlying beta cell development and expansion in normal human pancreatic development because it is likely that such mechanisms might also play a role in beta cell regeneration. The sensu strictiori definition of beta cells implies replacement of lost beta cell mass by new beta cells. In our discussion, however, we will use the term in a more general way, defining as regeneration the formation of new beta cells, whether or not a loss of beta cells has actually occurred. The potential mechanisms of beta cell regeneration in the human pancreas will be discussed in the second part of this review. In particular, we will analyze beta cell regeneration through proliferation of beta cells, neogenesis from non-beta cell precursors, and transdifferentiation from alpha cells. In the third part of this review, we will explore the arguments for and against the ability of the human pancreas to regenerate functional beta cells in the context of type 1 diabetes and in other pathological conditions.


Assuntos
Células Secretoras de Glucagon/citologia , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Pâncreas/citologia , Regeneração/fisiologia , Diferenciação Celular/fisiologia , Diabetes Mellitus Tipo 1/fisiopatologia , Células Secretoras de Glucagon/metabolismo , Humanos , Ductos Pancreáticos/citologia
14.
Curr Opin Endocrinol Diabetes Obes ; 16(4): 279-85, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19502978

RESUMO

PURPOSE OF REVIEW: Although studies of pancreata from type 1 diabetes (T1D) patients largely fell dormant for a period of decades, research efforts have recently been 'rekindled' in this area to address, using modern techniques, many unanswered questions related to the pathogenesis of this disease. RECENT FINDINGS: As historically noted, a pancreatic infiltrate commonly referred to as 'insulitis' is present at the symptomatic onset of T1D. Recent studies have further characterized this infiltrate both in terms of its cellular composition as well as the mechanisms that likely underlie beta cell death in T1D. In addition, the notion that the pancreas from T1D patients is completely devoid of insulin producing cells years after the onset of disease has been challenged, whereas the concepts of whether beta cell regeneration or replication are present have also been subject to much debate. Novel concepts regarding the rate and degree of beta cell loss throughout the natural history of the disease have also been put forward to aid in explaining the disorder's pathogenesis. SUMMARY: Although answers to many long-standing questions in T1D have recently been addressed, perhaps the main finding has been one supporting a disease of remarkable heterogeneity. However, additional lessons remain to be learned from the pancreas in T1D. Hence, attempts aimed at organizing the scientific community to address these issues are ongoing, particularly those from collaborative efforts, including the Belgium Organ Donor Consortium and the Network for Pancreatic Organ Donors with Diabetes (nPOD).


Assuntos
Diabetes Mellitus Tipo 1/patologia , Pâncreas/patologia , Glicemia/metabolismo , Diabetes Mellitus Tipo 1/prevenção & controle , Diabetes Mellitus Tipo 1/terapia , Humanos , Modelos Biológicos
15.
Novartis Found Symp ; 292: 85-94; discussion 94-8, 122-9, 202-3, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19203094

RESUMO

A wealth of data in animal models indicates that type 1A diabetes results from T cell-mediated specific destruction of islet beta cells. There is evidence for the NOD mouse model that insulin is the primary autoantigen and a specific insulin peptide B:9-23 is central to pathogenesis. It is also now possible to predict the development of type 1A (immune mediated) diabetes for the great majority of individuals with a combination of genetic, immunological and metabolic parameters. Such prediction is possible because of the chronic nature of the autoimmunity and loss of beta cell function that precedes the disease. Given the ability to predict type 1A diabetes trials at all stages of the disorder to prevent beta cell destruction are now possible.


Assuntos
Autoanticorpos/imunologia , Autoimunidade , Diabetes Mellitus Tipo 1/imunologia , Antígenos de Histocompatibilidade/imunologia , Insulina/imunologia , Ilhotas Pancreáticas/imunologia , Animais , Doença Crônica , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Predisposição Genética para Doença , Humanos , Ilhotas Pancreáticas/metabolismo , Redes e Vias Metabólicas/imunologia , Camundongos , Camundongos Endogâmicos NOD
16.
Cell Biochem Biophys ; 48(2-3): 177-82, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17709887

RESUMO

Even though there are numerous autoantigens for type 1 diabetes, current evidence suggests that a single autoantigen, namely insulin, is responsible for the key initiating event in autoimmunity. If a single autoantigen is necessary for triggering the autoimmune process, then antigen-specific therapy to block or delete the immune response against that autoantigen before epitope spreading occurs, may become a larger focus of future immunotherapeutic strategies. In this article, we review current literature regarding insulin as an autoantigen and potential approaches to deleting insulin-reactive T cells through the use of peptide vaccines and targeted T cell receptor immunizations.


Assuntos
Autoimunidade/imunologia , Diabetes Mellitus Tipo 1/imunologia , Insulina/imunologia , Ilhotas Pancreáticas/imunologia , Animais , Autoantígenos/imunologia , Diabetes Mellitus Tipo 1/terapia , Epitopos de Linfócito T/imunologia , Imunoterapia , Fragmentos de Peptídeos/imunologia , Linfócitos T/imunologia
18.
Gastroenterology ; 128(3): 728-41, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15765408

RESUMO

BACKGROUND & AIMS: The mammalian pancreas has a strong regenerative potential, but the origin of organ restoration is not clear, and it is not known to what degree such a process reflects pancreatic development. To define cell differentiation changes associated with pancreatic regeneration in adult mice, we compared regeneration following caerulein-induced pancreatitis to that of normal pancreatic development. METHODS: By performing comparative histology for adult and embryonic pancreatic markers in caerulein-treated and control pancreas, we addressed cellular proliferation and differentiation (amylase, DBA-agglutinin, insulin, glucagon, beta-catenin, E-cadherin, Pdx1, Nkx6.1, Notch1, Notch2, Jagged1, Jagged2, Hes1), hereby describing the kinetics of tissue restoration. RESULTS: We demonstrate that surviving pancreatic exocrine cells repress the terminal exocrine gene program and induce genes normally associated with undifferentiated pancreatic progenitor cells such as Pdx1, E-cadherin, beta-catenin, and Notch components, including Notch1 , Notch2 , and Jagged2 . Expression of the Notch target gene Hes1 provides evidence that Notch signaling is reactivated in dedifferentiated pancreatic cells. Although previous studies have suggested a process of acino-to-ductal transdifferentiation in pancreatic regeneration, we find no evidence to suggest that dedifferentiated cells acquire a ductal fate during this process. CONCLUSIONS: Pancreatic regeneration following chemically induced pancreatitis in the mouse occurs predominantly through acinar cell dedifferentiation, whereby a genetic program resembling embryonic pancreatic precursors is reinstated.


Assuntos
Pâncreas/embriologia , Pâncreas/fisiopatologia , Pancreatite/fisiopatologia , Regeneração , Animais , Ceruletídeo , Embrião de Mamíferos/metabolismo , Feminino , Regulação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos , Mitose , Pâncreas Exócrino/metabolismo , Pâncreas Exócrino/fisiopatologia , Pancreatite/induzido quimicamente , Pancreatite/genética , Pancreatite/metabolismo , Receptores Notch , Transdução de Sinais , Transativadores/metabolismo
19.
Immunol Rev ; 204: 232-49, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15790362

RESUMO

Type 1A diabetes is a chronic autoimmune disease usually preceded by a long prodrome during which autoantibodies to islet autoantigens are present. These antibodies are directed to a variety of antigens, but the best characterized are glutamic acid decarboxylase-65, insulinoma-associated antigen-2, and insulin. We hypothesize that the natural history of type 1A diabetes can be represented by several stages, starting from genetic susceptibility and ending in complete beta-cell destruction and overt diabetes. Type 1A diabetes probably results from a balance between genetic susceptibility and environmental influences. In both humans and animal models, the major determinants of the disease are genes within the major histocompatibility complex. The next best-characterized susceptibility locus is the insulin gene, the variable nucleotide tandem repeat locus. This gene affects the expression of insulin in the thymus and thus may play a role in the modulation of tolerance to this molecule. In a subset of genetically susceptible individuals, the activation of autoimmunity may be triggered by environmental factors such as viruses and/or diet. However, no conclusive association has been established between type 1A diabetes and specific environmental triggers. In this review, we provide evidence that insulin has a fundamental role in anti-islet autoimmunity.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/fisiopatologia , Animais , Autoanticorpos/imunologia , Autoantígenos/imunologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/terapia , Humanos , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia
20.
Clin Immunol ; 112(1): 54-65, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15207782

RESUMO

Signals transmitted by binding of Fas ligand (FasL) to the Fas receptor (CD95/Apo-1) have pleiotropic effects on cellular function that present opportunities for therapeutic applications. For example, depending on the circumstances, overexpression of FasL can enhance, prevent, or reverse growth of spontaneous or transplantable tumors. Furthermore, local administration of FasL into a single paw in susceptible mice protects from or reduces the severity of collagen-induced arthritis (CIA) in all paws. Here, we define mechanisms that mediate systemic protection induced by locally delivered FasL. Protection is not solely dependent on local interactions between Fas and FasL, but rather requires induction of a paradoxical inflammatory response that not only destroys Fas-resistant tumors, but also recruits motile, activated, Fas-bearing T cells that are Fas sensitive. We demonstrate by following the antigen-specific recruitment and subsequent termination of transgenic T cells that activated T cells, including autoreactive cells responsible for CIA, are eliminated within this inflammatory environment through the overexpressed FasL. The nature of the inflammatory response, which depends on the Fas ligand being cell bound and not soluble, and the magnitude of FasL expression within the inflammatory milieu are essential for this effect, as arthritogenic inflammation alone resulting from CIA induction is insufficient to ameliorate the disease or eliminate antigen-specific T cells, even upon systemic delivery of soluble FasL. These data show that gene delivery of membrane-bound FasL can effectively recruit and eliminate autoreactive T cells.


Assuntos
Artrite Experimental/prevenção & controle , Autoimunidade/efeitos dos fármacos , Glicoproteínas de Membrana/farmacologia , Linfócitos T/efeitos dos fármacos , Adenoviridae/genética , Transferência Adotiva , Animais , Artrite Experimental/imunologia , Artrite Experimental/patologia , Autoimunidade/imunologia , Morte Celular/imunologia , Citotoxicidade Imunológica , Proteína Ligante Fas , Feminino , Ativação Linfocitária , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Neoplasias Experimentais/imunologia , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...