Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Gut ; 2021 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-34330784

RESUMO

OBJECTIVE: The aggressive basal-like molecular subtype of pancreatic ductal adenocarcinoma (PDAC) harbours a ΔNp63 (p40) gene expression signature reminiscent of a basal cell type. Distinct from other epithelia with basal tumours, ΔNp63+ basal cells reportedly do not exist in the normal pancreas. DESIGN: We evaluated ΔNp63 expression in human pancreas, chronic pancreatitis (CP) and PDAC. We further studied in depth the non-cancerous tissue and developed a three-dimensional (3D) imaging protocol (FLIP-IT, Fluorescence Light sheet microscopic Imaging of Paraffin-embedded or Intact Tissue) to study formalin-fixed paraffin-embedded samples at single cell resolution. Pertinent mouse models and HPDE cells were analysed. RESULTS: In normal human pancreas, rare ΔNp63+ cells exist in ducts while their prevalence increases in CP and in a subset of PDAC. In non-cancer tissue, ΔNp63+ cells are atypical KRT19+ duct cells that overall lack SOX9 expression while they do express canonical basal markers and pertain to a niche of cells expressing gastrointestinal stem cell markers. 3D views show that the basal cells anchor on the basal membrane of normal medium to large ducts while in CP they exist in multilayer dome-like structures. In mice, ΔNp63 is not found in adult pancreas nor in selected models of CP or PDAC, but it is induced in organoids from larger Sox9low ducts. In HPDE, ΔNp63 supports a basal cell phenotype at the expense of a classical duct cell differentiation programme. CONCLUSION: In larger human pancreatic ducts, basal cells exist. ΔNp63 suppresses duct cell identity. These cells may play an important role in pancreatic disease, including PDAC ontogeny, but are not present in mouse models.

2.
Nucleic Acids Res ; 46(16): 8275-8298, 2018 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-29947794

RESUMO

CRISPR/Cas9 is an attractive platform to potentially correct dominant genetic diseases by gene editing with unprecedented precision. In the current proof-of-principle study, we explored the use of CRISPR/Cas9 for gene-editing in myotonic dystrophy type-1 (DM1), an autosomal-dominant muscle disorder, by excising the CTG-repeat expansion in the 3'-untranslated-region (UTR) of the human myotonic dystrophy protein kinase (DMPK) gene in DM1 patient-specific induced pluripotent stem cells (DM1-iPSC), DM1-iPSC-derived myogenic cells and DM1 patient-specific myoblasts. To eliminate the pathogenic gain-of-function mutant DMPK transcript, we designed a dual guide RNA based strategy that excises the CTG-repeat expansion with high efficiency, as confirmed by Southern blot and single molecule real-time (SMRT) sequencing. Correction efficiencies up to 90% could be attained in DM1-iPSC as confirmed at the clonal level, following ribonucleoprotein (RNP) transfection of CRISPR/Cas9 components without the need for selective enrichment. Expanded CTG repeat excision resulted in the disappearance of ribonuclear foci, a quintessential cellular phenotype of DM1, in the corrected DM1-iPSC, DM1-iPSC-derived myogenic cells and DM1 myoblasts. Consequently, the normal intracellular localization of the muscleblind-like splicing regulator 1 (MBNL1) was restored, resulting in the normalization of splicing pattern of SERCA1. This study validates the use of CRISPR/Cas9 for gene editing of repeat expansions.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes/métodos , Células-Tronco Pluripotentes Induzidas/metabolismo , Mioblastos/metabolismo , Distrofia Miotônica/genética , Expansão das Repetições de Trinucleotídeos/genética , Células Cultivadas , Criança , Feminino , Humanos , Pessoa de Meia-Idade , Desenvolvimento Muscular/genética , Distrofia Miotônica/metabolismo , Distrofia Miotônica/patologia
4.
Front Cell Dev Biol ; 10: 1041930, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36699012

RESUMO

The presence of sugar in the gut causes induction of SGLT1, the sodium/glucose cotransporter in intestinal epithelial cells (enterocytes), and this is accompanied by stimulation of sugar absorption. Sugar sensing was suggested to involve a G-protein coupled receptor and cAMP - protein kinase A signalling, but the sugar receptor has remained unknown. We show strong expression and co-localization with SGLT1 of the ß2-adrenergic receptor (ß 2-AR) at the enterocyte apical membrane and reveal its role in stimulating glucose uptake from the gut by the sodium/glucose-linked transporter, SGLT1. Upon heterologous expression in different reporter systems, the ß 2-AR responds to multiple sugars in the mM range, consistent with estimated gut sugar levels after a meal. Most adrenergic receptor antagonists inhibit sugar signaling, while some differentially inhibit epinephrine and sugar responses. However, sugars did not inhibit binding of I125-cyanopindolol, a ß 2-AR antagonist, to the ligand-binding site in cell-free membrane preparations. This suggests different but interdependent binding sites. Glucose uptake into everted sacs from rat intestine was stimulated by epinephrine and sugars in a ß 2-AR-dependent manner. STD-NMR confirmed direct physical binding of glucose to the ß 2-AR. Oral administration of glucose with a non-bioavailable ß 2-AR antagonist lowered the subsequent increase in blood glucose levels, confirming a role for enterocyte apical ß 2-ARs in stimulating gut glucose uptake, and suggesting enterocyte ß 2-AR as novel drug target in diabetic and obese patients. Future work will have to reveal how glucose sensing by enterocytes and neuroendocrine cells is connected, and whether ß 2-ARs mediate glucose sensing also in other tissues.

5.
Virchows Arch ; 478(6): 1209-1214, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32833039

RESUMO

Insulitis is a characteristic inflammatory lesion consisting of immune cell infiltrates around and within the pancreatic islets of patients with recent-onset type 1 diabetes (T1D). The infiltration is typically mild, both in terms of the number of infiltrating cells and the number of islets affected. Here, we present an unusual histopathological case study of a 66-year-old female patient with long-standing T1D, insulitis, and islet-associated lymphoid tissue. Most islets in the head of the pancreas of this patient were insulin-deficient, whereas the islets in the tail appeared normal. Insulitis was present in 0.84% of the insulin-containing islets and three islets had large lymphocytic infiltrates resembling tertiary lymphoid structures (TLS). Of note, this is the first description of potential TLS in the endocrine pancreas of a patient with T1D. Their association with a marked residual beta cell mass is of interest and may hint at new insights into disease progression and regulation of autoimmunity.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Ilhotas Pancreáticas/patologia , Linfonodos/patologia , Idoso , Autoimunidade/imunologia , Diabetes Mellitus Tipo 1/complicações , Progressão da Doença , Feminino , Humanos , Insulina/metabolismo , Ilhotas Pancreáticas/imunologia , Linfonodos/imunologia
6.
Virchows Arch ; 479(2): 295-304, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33594586

RESUMO

Autoantibodies against islet cell antigens are routinely used to identify subjects at increased risk of symptomatic type 1 diabetes, but their relation to the intra-islet pathogenetic process that leads to positivity for these markers is poorly understood. We screened 556 non-diabetic organ donors (3 months to 24 years) for five different autoantibodies and found positivity in 27 subjects, 25 single- and two double autoantibody-positive donors. Histopathological screening of pancreatic tissue samples showed lesion characteristic for recent-onset type 1 diabetes in the two organ donors with a high-risk profile, due to their positivity for multiple autoantibodies and HLA-inferred risk. Inflammatory infiltrates (insulitis) were found in a small fraction of islets (<5%) and consisted predominantly of CD3+CD8+ T-cells. Islets with insulitis were found in close proximity to islets devoid of insulin-positivity; such pseudo-atrophic islets were present in multiple small foci scattered throughout the pancreatic tissue or were found to be distributed with a lobular pattern. Relative beta cell area in both single and multiple autoantibody-positive donors was comparable to that in autoantibody-negative controls. In conclusion, in organ donors under age 25 years, insulitis and pseudo-atrophic islets were restricted to multiple autoantibody-positive individuals allegedly at high risk of developing symptomatic type 1 diabetes, in line with reports in older age groups. These observations may give further insight into the early pathogenetic events that may culminate in clinically overt disease.


Assuntos
Antígenos/imunologia , Autoanticorpos/sangue , Linfócitos T CD8-Positivos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Células Secretoras de Insulina/imunologia , Transplante das Ilhotas Pancreáticas , Doadores de Tecidos , Adolescente , Fatores Etários , Biomarcadores/sangue , Estudos de Casos e Controles , Proliferação de Células , Criança , Pré-Escolar , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/patologia , Seleção do Doador , Feminino , Humanos , Lactente , Células Secretoras de Insulina/patologia , Masculino , Adulto Jovem
8.
Adv Exp Med Biol ; 654: 1-19, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20217491

RESUMO

Human islets of Langerhans are complex micro-organs responsible for maintaining glucose homeostasis. Islets contain five different endocrine cell types, which react to changes in plasma nutrient levels with the release of a carefully balanced mixture of islet hormones into the portal vein. Each endocrine cell type is characterized by its own typical secretory granule morphology, different peptide hormone content, and specific endocrine, paracrine, and neuronal interactions. During development, a cascade of transcription factors determines the formation of the endocrine pancreas and its constituting islet cell types. Differences in ontogeny between the ventrally derived head section and the dorsally derived head, body, and tail section are responsible for differences in innervation, blood supply, and endocrine composition. Islet cells show a close topographical relationship to the islet vasculature, and are supplied with a five to tenfold higher blood flow than the exocrine compartment. Islet microanatomy is disturbed in patients with type 1 diabetes, with a marked reduction in beta-cell content and the presence of inflammatory infiltrates. Histopathological lesions in type 2 diabetes are less pathognomonic with a more limited reduction in beta-cell content and occasional deposition of amyloid in the islet interstitial space.


Assuntos
Ilhotas Pancreáticas/anatomia & histologia , Ilhotas Pancreáticas/fisiologia , Amiloide/metabolismo , Animais , Autoimunidade , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 2/patologia , Glucose/metabolismo , Homeostase , Humanos , Inflamação , Células Secretoras de Insulina/citologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/ultraestrutura , Modelos Anatômicos , Modelos Biológicos
9.
Diabetes ; 69(3): 401-412, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31843955

RESUMO

M2 macrophages play an important role in tissue repair and regeneration. They have also been found to modulate ß-cell replication in mouse models of pancreatic injury and disease. We previously reported that ß-cell replication is strongly increased in a subgroup of human organ donors characterized by prolonged duration of stay in an intensive care unit (ICU) and increased number of leukocytes in the pancreatic tissue. In the present study we investigated the relationship between duration of stay in the ICU, M2 macrophages, vascularization, and pancreatic cell replication. Pancreatic organs from 50 donors without diabetes with different durations of stay in the ICU were analyzed by immunostaining and digital image analysis. The number of CD68+CD206+ M2 macrophages increased three- to sixfold from ≥6 days' duration of stay in the ICU onwards. This was accompanied by a threefold increased vascular density and a four- to ninefold increase in pancreatic cells positive for the replication marker Ki67. A strong correlation was observed between the number of M2 macrophages and ß-cell replication. These results show that a prolonged duration of stay in the ICU is associated with an increased M2 macrophage number, increased vascular density, and an overall increase in replication of all pancreatic cell types. Our data show evidence of marked levels of tissue repair in the human donor pancreas.


Assuntos
Proliferação de Células/fisiologia , Unidades de Terapia Intensiva , Tempo de Internação , Macrófagos/patologia , Pâncreas/fisiologia , Regeneração/fisiologia , Doadores de Tecidos , Adolescente , Adulto , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Feminino , Humanos , Antígeno Ki-67/metabolismo , Lectinas Tipo C/metabolismo , Macrófagos/metabolismo , Masculino , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Pessoa de Meia-Idade , Neovascularização Fisiológica/fisiologia , Pâncreas/metabolismo , Pâncreas/patologia , Receptores de Superfície Celular/metabolismo , Adulto Jovem
10.
Hum Reprod ; 24(5): 1085-91, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19176542

RESUMO

BACKGROUND: Previously, advanced endometrial maturation on the day of oocyte retrieval in GnRH antagonist and -agonist IVF cycles was observed. In these cycles, endometrial advancement exceeding 3 days between the histological dating and the cycle day never resulted in an ongoing clinical pregnancy. In this study, the gene expression of human endometrium on the day of oocyte retrieval in GnRH antagonist/rec-FSH cycles was analyzed, in correlation with the morphological dating. METHODS: Biopsies were taken on the day of oocyte retrieval in 47 patients with 1 or 2 embryos replaced on Day 3 in the same cycle. Endometrial dating was performed according to Noyes' criteria. Biopsies from 11 patients were analyzed for gene expression with the Affymetrix HG U133 Plus 2 microarray. Data analysis, clustering and pathway analysis were performed with GCOS, GeneSpring 7.3 and Ingenuity, respectively. RESULTS: According to Noyes' criteria, all endometria taken on the day of oocyte retrieval showed an advanced maturation, ranging from +d2 to +d4. The patients with a subsequent clinical pregnancy all showed a histological dating corresponding to +d2 or +d3. When comparing endometria +d2-3 to +d4, the microarray results showed a differential expression of 2550 probe sets. Significantly up-regulated genes were SERPINB6, FOXO3A, SOX17 and CDC42. Down-regulated genes of interest were NRP1, HOXA10 and OSF2. Principal component analysis and hierarchical clustering demonstrated two distinct clusters. CONCLUSIONS: In stimulated cycles, endometrial gene expression on the day of oocyte retrieval discriminates between women with and without histologically advanced endometrial maturation exceeding 3 days and supports histological dating results by Noyes' criteria.


Assuntos
Endométrio/efeitos dos fármacos , Hormônio Foliculoestimulante/farmacologia , Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Antagonistas de Hormônios/farmacologia , Indução da Ovulação , Adulto , Análise por Conglomerados , Endométrio/metabolismo , Feminino , Perfilação da Expressão Gênica , Hormônio Liberador de Gonadotropina/farmacologia , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Recuperação de Oócitos , Gravidez , Resultado da Gravidez , Proteínas Recombinantes/farmacologia
11.
J Cell Biol ; 159(2): 303-12, 2002 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-12403815

RESUMO

Regulatory proteins have been identified in embryonic development of the endocrine pancreas. It is unknown whether these factors can also play a role in the formation of pancreatic endocrine cells from postnatal nonendocrine cells. The present study demonstrates that adult human pancreatic duct cells can be converted into insulin-expressing cells after ectopic, adenovirus-mediated expression of the class B basic helix-loop-helix factor neurogenin 3 (ngn3), which is a critical factor in embryogenesis of the mouse endocrine pancreas. Infection with adenovirus ngn3 (Adngn3) induced gene and/or protein expression of NeuroD/beta2, Pax4, Nkx2.2, Pax6, and Nkx6.1, all known to be essential for beta-cell differentiation in mouse embryos. Expression of ngn3 in adult human duct cells induced Notch ligands Dll1 and Dll4 and neuroendocrine- and beta-cell-specific markers: it increased the percentage of synaptophysin- and insulin-positive cells 15-fold in ngn3-infected versus control cells. Infection with NeuroD/beta2 (a downstream target of ngn3) induced similar effects. These data indicate that the Delta-Notch pathway, which controls embryonic development of the mouse endocrine pancreas, can also operate in adult human duct cells driving them to a neuroendocrine phenotype with the formation of insulin-expressing cells.


Assuntos
Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/fisiologia , Proteínas do Tecido Nervoso/genética , Ductos Pancreáticos/citologia , Ductos Pancreáticos/fisiologia , Adulto , Fatores Etários , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Biomarcadores , Diferenciação Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodomínio , Humanos , Insulina/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Ilhotas Pancreáticas/embriologia , Proteínas de Membrana/genética , Proteínas Nucleares , Ductos Pancreáticos/embriologia , Fenótipo , Receptores Notch , Fatores de Transcrição , Transcrição Gênica/fisiologia
12.
Novartis Found Symp ; 292: 19-24; discussion 24-31, 122-9, 202-3, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19203090

RESUMO

Type 1 diabetes is often considered as a disease where more than 90% of the beta cells have been destroyed at clinical onset and where beta cell antigen-driven autoimmune reactivities progressively destroy remaining beta cells as well as newly formed or implanted beta cells. This view will be evaluated in light of histological observations in the pancreas of type 1 diabetic patients and of antibody-positive non-diabetic organ donors.


Assuntos
Diabetes Mellitus Tipo 1/patologia , Células Secretoras de Insulina/patologia , Autoanticorpos/imunologia , Autoanticorpos/metabolismo , Sobrevivência Celular , Diabetes Mellitus Tipo 1/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/metabolismo , Linfócitos/imunologia
13.
BMC Cancer ; 8: 3, 2008 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-18182111

RESUMO

BACKGROUND: The status of the EGFR and HER2-neu genes has not been fully defined in ovarian cancer. An integrated analysis of both genes could help define the proportion of patients that would potentially benefit from targeted therapies. METHODS: We determined the tumour mutation status of the entire tyrosine kinase (TK) domain of the EGFR and HER2-neu genes in a cohort of 52 patients with invasive epithelial ovarian cancer as well as the gene copy number and protein expression of both genes in 31 of these patients by DGGE and direct sequecing, immunohistochemistry and Fluorescent in Situ Hybridisation (FISH). RESULTS: The EGFR was expressed in 59% of the cases, with a 2+/3+ staining intensity in 38%. HER2-neu expression was found in 35%, with a 2/3+ staining in 18%. No mutations were found in exons 18-24 of the TK domains of EGFR and HER2-neu. High polysomy of the EGFR gene was observed in 13% of the invasive epthelial cancers and amplification of the HER2-neu gene was found in 10% and correlated with a high expression level by immunohistochemistry.Mutations within the tyrosine kinase domain were not found in the entire TK domain of both genes, but have been found in very rare cases by others. CONCLUSION: Genomic alteration of the HER2-neu and EGFR genes is frequent (25%) in ovarian cancer. EGFR/HER2-neu targeted therapies should be investigated prospectively and specifically in that subset of patients.


Assuntos
Carcinoma/genética , Regulação Neoplásica da Expressão Gênica , Genes erbB-1 , Genes erbB-2 , Invasividade Neoplásica/patologia , Neoplasias Ovarianas/genética , Idoso , Bélgica , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Biópsia por Agulha , Carcinoma/mortalidade , Carcinoma/patologia , Análise Mutacional de DNA , Progressão da Doença , Feminino , Genômica , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Pessoa de Meia-Idade , Neoplasias Ovarianas/mortalidade , Neoplasias Ovarianas/patologia , Valor Preditivo dos Testes , Probabilidade , Prognóstico , Estudos Retrospectivos , Medição de Risco , Estudos de Amostragem , Sensibilidade e Especificidade , Análise de Sobrevida
14.
Diabetes ; 55(1): 78-85, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16380479

RESUMO

Use of sulfonylureas in diabetes treatment is based on their insulin-releasing effect on pancreatic beta-cells. Prolonged action is known to degranulate beta-cells, but functional consequences have not been examined at the cellular level. This study investigates influences of in vivo (48-h) and in vitro (24-h) glibenclamide treatment on the functional state of the beta-cell population. Both conditions decreased cellular insulin content by >50% and caused an elevated basal insulin biosynthetic activity that was maintained for at least 24 h after drug removal. Glibenclamide stimulation of basal insulin synthesis was not achieved after a 2-h exposure; it required a calcium-dependent translational activity and involved an increase in the percent activated beta-cells (50% after glibenclamide pretreatment vs. 8% in control cells). The glibenclamide-activated beta-cell subpopulation corresponded to the degranulated beta-cell subpopulation that was isolated by fluorescence-activated cell sorter on the basis of lower cellular sideward scatter. Glibenclamide pretreatment did not alter cellular rates of glucose oxidation but sensitized beta-cells to glucose-induced changes in metabolic redox and insulin synthesis and release. In conclusion, chronic exposure to glibenclamide results in degranulation of a subpopulation of beta-cells, which maintain an elevated protein and insulin synthetic activity irrespective of the presence of the drug and of glucose. Our study demonstrates that the in situ beta-cell population also exhibits a functional heterogeneity that can vary with drug treatment. Glibenclamide induces degranulated beta-cells with a sustained elevated basal activity that might increase the risk for hypoglycemic episodes.


Assuntos
Glibureto/farmacologia , Hipoglicemiantes/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Insulina/biossíntese , Animais , Cálcio/metabolismo , Degranulação Celular/efeitos dos fármacos , Esquema de Medicação , Células Secretoras de Insulina/citologia , Masculino , Ratos , Ratos Wistar
15.
Diabetes ; 66(2): 426-436, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27864307

RESUMO

ß-Cell failure in type 2 diabetes (T2D) was recently proposed to involve dedifferentiation of ß-cells and ectopic expression of other islet hormones, including somatostatin and glucagon. Here we show that gastrin, a stomach hormone typically expressed in the pancreas only during embryogenesis, is expressed in islets of diabetic rodents and humans with T2D. Although gastrin in mice is expressed in insulin+ cells, gastrin expression in humans with T2D occurs in both insulin+ and somatostatin+ cells. Genetic lineage tracing in mice indicates that gastrin expression is turned on in a subset of differentiated ß-cells after exposure to severe hyperglycemia. Gastrin expression in adult ß-cells does not involve the endocrine progenitor cell regulator neurogenin3 but requires membrane depolarization, calcium influx, and calcineurin signaling. In vivo and in vitro experiments show that gastrin expression is rapidly eliminated upon exposure of ß-cells to normal glucose levels. These results reveal the fetal hormone gastrin as a novel marker for reversible human ß-cell reprogramming in diabetes.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Gastrinas/metabolismo , Células Secretoras de Insulina/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Estudos de Casos e Controles , Diabetes Mellitus/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Gerbillinae , Humanos , Imuno-Histoquímica , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Células Secretoras de Somatostatina/metabolismo , Células-Tronco/metabolismo
16.
Cell Transplant ; 26(1): 1-9, 2017 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-27729094

RESUMO

Transplantation of islet allografts into type 1 diabetic recipients usually requires multiple pancreas donors to achieve insulin independence. This adds to the challenges of immunological monitoring of islet transplantation currently relying on surrogate immune markers in peripheral blood. We investigated donor origin and infiltration of islets transplanted in the liver of a T1D patient who died of hemorrhagic stroke 4 months after successful transplantation with two intraportal islet grafts combining six donors. Immunohistological staining for donor HLA using a unique panel of human monoclonal HLA-specific alloantibodies was performed on liver cryosections after validation on cryopreserved kidney, liver, and pancreas and compared with auto- and alloreactive T-cell immunity in peripheral blood. HLA-specific staining intensity and signal-to-noise ratio varied between tissues from very strong on kidney glomeruli, less in liver, kidney tubuli, and endocrine pancreas to least in exocrine pancreas, complicating the staining of inflamed islets in an HLA-disparate liver. Nonetheless, five islets from different liver lobes could be attributed to donors 1, 2, and 5 by staining patterns with multiple HLA types. All islets showed infiltration with CD8+ cytotoxic T cells that was mirrored by progressive alloreactive responses in peripheral blood mononuclear cells (PBMCs) to donors 1, 2, and 5 after transplantation. Stably low rates of peripheral islet autoreactive T-cell responses after islet infusion fit with a complete HLA mismatch between grafts and recipient and exclude the possibility that the islet-infiltrating CD8 T cells were autoreactive. HLA-specific immunohistochemistry can identify donor origin in situ and differentiate graft dysfunction and immunological destruction.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/cirurgia , Transplante das Ilhotas Pancreáticas/imunologia , Doadores de Tecidos , Autoimunidade/imunologia , Linfócitos T CD8-Positivos/metabolismo , Diabetes Mellitus Tipo 1/imunologia , Feminino , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Fígado/metabolismo , Pessoa de Meia-Idade , Pâncreas/imunologia , Pâncreas/metabolismo , Transplante Homólogo
17.
PLoS One ; 12(8): e0181651, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28792951

RESUMO

The specific phenotype of mature differentiated beta cells not only depends on the specific presence of genes that allow beta cell function but also on the selective absence of housekeeping genes ("disallowed genes") that would interfere with this function. Recent studies have shown that both histone modifications and DNA methylation via the de novo methyltransferase DNMT3A are involved in repression of disallowed genes in neonatal beta cells when these cells acquire their mature phenotype. It is unknown, however, if the environmental influence of advanced age, pregnancy and the metabolic stress of high fat diet or diabetes could alter the repression of disallowed genes in beta cells. In the present study, we show that islet disallowed genes-which are also deeply repressed in FACS-purified beta cells-remain deeply repressed in animals of advanced age and in pregnant females. Moreover, the stability of this repression was correlated with strong and stable histone repression marks that persisted in islets isolated from 2 year old mice and with overall high expression of Dnmt3a in islets. Furthermore, repression of disallowed genes was unaffected by the metabolic stress of high fat diet. However, repression of about half of the disallowed genes was weakened in 16 week-old diabetic db/db mice. In conclusion, we show that the disallowed status of islet genes is stable under physiological challenging conditions (advanced age, pregnancy, high fat diet) but partially lost in islets from diabetic animals.


Assuntos
Envelhecimento/fisiologia , Metilação de DNA/genética , Diabetes Mellitus/metabolismo , Dieta Hiperlipídica , Código das Histonas/genética , Células Secretoras de Insulina/metabolismo , Estresse Fisiológico/fisiologia , Animais , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Metiltransferase 3A , Feminino , Teste de Tolerância a Glucose , Insulina/metabolismo , Células Secretoras de Insulina/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Gravidez
18.
Diabetes ; 54(12): 3387-94, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16306353

RESUMO

The development of islet cell transplantation as a cure for diabetes is limited by the shortage of human donor organs. Moreover, currently used grafts exhibit a marginal beta-cell mass with an apparently low capacity for beta-cell renewal and growth. Although duct-associated nonendocrine cells have often been suggested as a potential source for beta-cell production, recent work in mice has demonstrated the role of beta-cells in postnatal growth of the pancreatic beta-cell mass. The present study investigated whether the beta-cell mass can grow in implants that are virtually devoid of nonendocrine cells. Endocrine islet cells were purified from prenatal porcine pancreases (gestation >110 days) and implanted under the kidney capsule of nude mice. beta-Cells initially presented with signs of immaturity: small size, low insulin content, undetectable C-peptide release, and an inability to correct hyperglycemia. They exhibited a proliferative activity that was highest during posttransplant week 1 (2.6 and 5% bromodeoxyuridine [BrdU]-positive beta-cells 4 and 72 h posttransplant) and then decreased over 20 weeks to rates measured in the pancreas (0.2% BrdU-positive cells). beta-Cell proliferation in implants first compensated for beta-cell loss during posttransplant week 1 and then increased the beta-cell number fourfold between posttransplant weeks 1 and 20. Rates of alpha-cell proliferation were only shortly and moderately increased, which explained the shift in cellular composition of the implant (beta-cell 40 vs. 90% and alpha-cell 40 vs. 7% at the start and posttransplant week 20, respectively). beta-Cells progressively matured during the 20 weeks after transplantation, with a twofold increase in cell volume, a sixfold increase in cellular insulin content, plasma C-peptide levels of 1-2 ng/ml, and an ability to correct diabetes. They became structurally organized as homogenous clusters with their secretory vesicles polarized toward fenestrated capillaries. We concluded that the immature beta-cell phenotype provides grafts with a marked potential for beta-cell growth and differentiation and hence may have a potential role in curing diabetes. Cells with this phenotype can be isolated from prenatal organs; their presence in postnatal organs needs to be investigated.


Assuntos
Transplante das Ilhotas Pancreáticas/fisiologia , Pâncreas/embriologia , Animais , Animais Recém-Nascidos , Glicemia/metabolismo , Peptídeo C/sangue , Divisão Celular , Glucagon/análise , Insulina/análise , Transplante das Ilhotas Pancreáticas/patologia , Camundongos , Camundongos Nus , Suínos , Sinaptofisina/análise , Fatores de Tempo , Transplante Heterólogo
19.
Diabetes ; 54(7): 2132-42, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15983215

RESUMO

Cellular redox state is an important metabolic variable, influencing many aspects of cell function like growth, apoptosis, and reductive biosynthesis. In this report, we identify NADPH as a candidate signaling molecule for exocytosis in neuroendocrine cells. In pancreatic beta-cells, glucose acutely raised the NADPH-to-NADP+ ratio and stimulated insulin release in parallel. Furthermore, intracellular addition of NADPH directly stimulated exocytosis of insulin granules. Effects of NADPH on exocytosis are proposed to be mediated by the redox proteins glutaredoxin (GRX) and thioredoxin (TRX) on the basis of the following evidence: 1) Expression of GRX mRNA is very high in beta-cells compared with other studied tissues, and GRX protein expression is high in islets and in brain; 2) GRX and TRX are localized in distinct microdomains in the cytosol of beta-cells; and 3) microinjection of recombinant GRX potentiated effects of NADPH on exocytosis, whereas TRX antagonized the NADPH effect. We propose that the NADPH/GRX/TRX redox regulation mediates a novel signaling pathway of nutrient-induced insulin secretion.


Assuntos
Exocitose/fisiologia , Insulina/metabolismo , Ilhotas Pancreáticas/fisiologia , NADP/metabolismo , Oxirredutases/genética , Tiorredoxinas/genética , Animais , Sequência de Bases , Primers do DNA , Exocitose/efeitos dos fármacos , Glucose/farmacologia , Glutarredoxinas , Homeostase , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , RNA Mensageiro/genética , Ratos
20.
Virchows Arch ; 446(3): 232-8, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15791482

RESUMO

Amyloid-containing (A+) islets are characteristic for type-2 diabetes (T2D), but their abundance seems variable among patients. It is unclear whether the distribution of A+ islets follows a certain pattern or occurs randomly throughout the pancreatic organ. We investigated the topography of A+ islets in eight pancreata of T2D patients and eight sex- and age-matched non-diabetic subjects. Transversal sections of head, body and tail segments were stained with synaptophysin combined with Congo red to map/quantify islet tissue and amyloid. In the eight T2D pancreata, the overall percentage of A+ islets varied from 4% to 85%. Further analysis in body and tail indicated that peripheral regions exhibited higher percentages of A+ islets than central regions (averages of, respectively, 30% and 17%, P<0.05). Non-diabetic control pancreata also exhibited A+ islets, albeit at a 25-fold lower frequency; a tendency towards higher percentage of A+ islets in peripheral versus central regions was also observed. The higher percentage A+ islets in peripheral regions was associated with a higher density and relative islet over exocrine surface area. These observations on heterogeneity in abundance and distribution of A+ islets need consideration when sampling tissue for studies on human islet amyloidosis. The present methodology allows us to further investigate the susceptibility to amyloidosis of islets in peripheral regions of the pancreas.


Assuntos
Amiloide/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Masculino , Polipeptídeo Pancreático/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA