• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 154
  • 89
  • 29
  • 14
  • 10
  • 4
  • 3
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 1
  • Tagged with
  • 321
  • 154
  • 120
  • 106
  • 73
  • 67
  • 61
  • 60
  • 58
  • 58
  • 42
  • 41
  • 37
  • 37
  • 35
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
301

The Genetic Basis of Resistance to Transplantation Tolerance Induced by Costimulation Blockade in NOD Mice: a Dissertation

Pearson, Todd 17 March 2003 (has links)
The NOD mouse is a widely studied model of type 1 diabetes. The loss of self-tolerance leading to autoimmune diabetes in NOD mice involves at least 27 genetic loci. Curing type I diabetes in mice and humans by islet transplantation requires overcoming both allorejection and recurrent autoimmunity. This has been achieved with systemic immunosuppression, but tolerance induction would be preferable. In addition to their genetic defects in self-tolerance, NOD mice resist peripheral transplantation tolerance induced by costimulation blockade using donor-specific transfusion and anti-CDl54 antibody. Failure has been attributed to the underlying autoimmunity, assuming that autoimmunity and resistance to transplantation tolerance have a common basis. Hypothesizing that these two abnormalities might be related, we investigated whether they had a common genetic basis. Diabetes-resistant NOD and C57BL/6 stocks congenic for various reciprocally introduced Idd loci were assessed for their ability to be tolerized. Surprisingly, in NOD congenic mice that are almost completely protected from diabetes, costimulation blockade failed to prolong skin allograft survival. In reciprocal C57BL/6 congenic mice with NOD-derived Idd loci, skin allograft survival was readily prolonged by costimulation blockade. Unexpectedly, we observed that (NOD x C57BL/6)F1 mice, which have no diabetes, nonetheless resist induction of tolerance to skin allografts. Further analyses revealed that the F1 mice shared the dendritic cell maturation defects and abnormal CD4+ T cell responses of the NOD but had lost its defects in macrophage maturation and NK cell activity. Finally, using a genome wide scan approach, we have identified four suggestive markers in the mouse genome that control the survival of skin allografts following DST and anti-CD154 mAb therapy. We suggest that mechanisms controlling autoimmunity and transplantation tolerance in NOD mice are not completely overlapping and are potentially distinct, or that the genetic threshold for normalizing the transplantation tolerance defect is higher than that for preventing autoimmune diabetes. We conclude that resistance to allograft tolerance induction in the NOD mouse is not a direct consequence of overt autoimmunity and that autoimmunity and resistance to costimulation blockade-induced transplantation tolerance phenotypes in NOD mice are not under identical genetic control.
302

Diagnostický příspěvek k hodnocení intervenčních modelů léčby diabetu mellitu 1. typu / Diagnostic contribution to the evaluation of intervention models in the treatment of type 1 diabetes

Zacharovová, Klára January 2012 (has links)
During treatment of diabetes mellitus by immunointervention or transplantation, it is necessary to monitor the markers of immune destruction or rejection of surviving insulin producing cells. An aim of this thesis is to improve the possibilities of following autoimmunity and to detect the survival of transplanted pancreatic islet in vivo. Partial aims included vitality testing of isolated islets for transplantation by measurement of respiration activity, observing the process of in vitro labeling of isolated islets with superparamagnetic iron oxide (SPIO) contrast agent for subsequent magnetic resonance imaging (MRI) of islets and observing SPIO particles transport after transplantation. We also studied a new dual paramagnetic contrast agent combined with fluorescein intended for identification of the MRI contrast agent in samples for histology. Further, we assessed autoimmune reaction by evaluation of cytokine response to specific stimulation with auto-antigens. We tried to affect beta-cells destruction by polyclonal anti- thymocyte antibodies in a mouse experimental model. A new method of the islet respiration measurement correlated with other methods of islet quality testing and it was suggested as a diagnostic test before clinical transplantation. Results obtained studying the intercellular...
303

Ovlivnění funkce ischemicky poškozených orgánů použitím perfluorocarbonu (PFC) jako konzervačního roztoku při experimentální transplantaci pankreatu, ledviny a Langerhansových ostrůvků / Posttransplant function of ischemically impaired organs (pancreas, kidney, islets) preserved by perfluorocarbon (PFC)

Marada, Tomáš January 2013 (has links)
(English) Perfluorocarbons (PFC) are hydrocarbons in which some or all of the hydrogen atoms are replaced with fluorine. PFC have a very high capacity for dissolving oxygen. They are chemically and biologically inert. The most successful clinical application of PFC is the "two-layer method" for pancreas preservation before islet isolation. The two-layer organ preservation method (TLM) is based on oxygenated perfluorocarbon overlaid with University of Wisconsin (UW) solution. In experiment it has been successfully used for heart and intestine transplantation. We tested whether this technique would prevent tissue damage and improve results of kidney, pancreas and islets of Langerhans transplantation with prolonged ischemia time in an experimental model of syngenic rats. In kidney and islets of Langerhanse transplantation model we used TLM preservation method. In pancreas transplantation model we used perfluorohexyloctane (PFH) as a new generation of less lipophilic PFC. 1. Kidneys were stored for 24 hours either in UW solution (n = 16), with TLM (n = 16) or transplanted immediately (control group, n = 12). In half of the animals, survival was observed and in the other animals grafts were procured for semiquantitative histological scoring and TUNEL apoptosis assessment 24 h after transplantation....
304

O efeito pró-apoptótico de oligômeros da amilina humana não é potencializado pela lipotoxicidade em ilhotas pancreáticas de rato em cultura / The pro-apoptotic effect of human amylin oligomers is not potentiated by lipotoxicity in rat pancreatic islets in culture

Érika Rodrigues de Oliveira 25 July 2012 (has links)
O depósito de amilina é um achado histopatológico frequente em pacientes portadores de diabetes mellitus tipo 2 (DM 2) e parece estar relacionado à disfunção da célula beta pancreática característica desta doença. Um estudo previamente desenvolvido em nosso laboratório verificou que oligômeros de amilina humana provocam diminuição na expressão do mRNA do gene que codifica o receptor do hormônio incretínico peptídeo insulinotrópico dependente de glicose (Gipr) e aumento do índice de apoptose em ilhotas pancreáticas de rato mantidas em cultura. Considerando o importante papel do depósito amilóide e das incretinas na fisiopatologia do DM 2, os objetivos deste trabalho foram investigar (1) o efeito da amilina humana sobre a expressão dos receptores de incretinas e (2) a modulação de seu efeito tóxico por outras condições concomitantes presentes no DM, como a lipotoxicidade e os produtos finais de glicação avançada (AGEs). Para isto, foi realizada a avaliação da expressão do mRNA dos genes Gipr e Glp1r (receptor do peptídeo semelhante ao glucagon) por PCR em tempo real em ilhotas expostas apenas aos oligômeros de amilina humana (10 M) por 4 e 8 h e em ilhotas expostas aos oligômeros e ao palmitato (0,5 mM) por 24 e 48 h; avaliação da expressão das proteínas GIPR e GLP1R por Western blot em ilhotas tratadas com oligômeros de amilina por 12 h; e avaliação do índice de apoptose pela quantificação da atividade de caspase 3 em ilhotas tratadas com oligômeros de amilina isoladamente, ou na presença de palmitato (0,5mM) por 48 h ou 5 mg/ml de albumina glicada (AlbGAD) por 72 h. A amilina não provocou alteração na expressão dos genes Gipr e Glp1r após 4 h de exposição. Após 8 e 24 h de tratamento, os oligômeros modularam negativamente a expressão destes genes. Entretanto, o tratamento das ilhotas com amilina por 48 h resultou no aumento da expressão do mRNA dos receptores de incretinas. O tratamento simultâneo com palmitato não alterou o efeito modulatório da amilina sobre a expressão dos genes Gipr e Glp1r após 24 e 48 h. A exposição das ilhotas aos oligômeros de amilina por 12 h não causou alteração na expressão das proteínas GIPR e GLP1R. A lipotoxicidade e a albumina glicada não aumentaram o efeito pró-apoptótico da amilina sobre as ilhotas pancreáticas. Em conclusão, a redução na expressão gênica dos receptores de incretinas em ilhotas pancreáticas de rato expostas aos oligômeros de amilina, que poderia indicar um mecanismo adicional pelo qual a amilina exerceria seu efeito deletério sobre células beta, diminuindo o efeito insulinotrópico induzido pelas incretinas em pacientes com DM 2, não foi constatada em relação à expressão protéica de GIPR e GLP1R no período de tempo estudado. O aumento na expressão do mRNA destes receptores provocado pela amilina após 48 horas de incubação poderia ser um mecanismo de compensação das células frente aos efeitos tóxicos dos oligômeros de amilina. O efeito próapoptótico da amilina humana sobre as células beta não parece ser potencializado pela lipotoxicidade ou por AGEs / The amyloid deposit is a common histopathological feature in patients with type 2 diabetes mellitus (T2DM) and it seems to be related to the pancreatic beta cell dysfunction characteristic of this disease. A study previously developed in our laboratory found that human amylin oligomers decrease mRNA expression of the glucose-dependent insulinotropic polypeptide receptor (Gipr) and increase apoptosis rate in rat pancreatic islets maintained in culture. Considering the important role of the amyloid deposition and of incretins in the pathophysiology of T2DM, the aims of the present study were to investigate (1) the effect of human amylin on the expression of incretin receptors and (2) the modulation of amylin toxicity by other concomitant conditions present in T2DM, as lipotoxicity and advanced glycation end products (AGEs). The evaluation of mRNA expression of Gipr and Glp1r (glucagonlike peptide -1 receptor) was performed by real time PCR in islets exposed only to human amylin oligomers (10 M) for 4 and 8 h, and in islets exposed to human amylin and palmitate (0,5 mM) for 24 and 48 h; GIPR and GLP1R protein expression was assessed by Western blot in islets treated with amylin oligomers by 12 h; apoptosis rate was evaluated by measuring caspase 3 activity in islets treated with amylin alone or combined to palmitate (0,5 mM) for 48 h or 5 mg/mL of glycated albumin (AlbGAD) for 72 h. Amylin did not affect the expression of Gipr and Glp1r mRNA following 4 h of exposure. Eight and 24 h after treatment, amylin negatively modulated the expression of these genes. However, treatment of the islets for 48 h with amylin elicited an increase in mRNA expression of both incretin receptors. The simultaneous treatment with palmitate did not change the effects of amylin on the expression of Gipr and Glp1r mRNA after 24 and 48 h. Exposure of islets to amylin for 12 h caused no change in GIPR and GLP1R protein expression. Lipotoxocity and glycated albumin did not increase the pro-apoptotic effect of amylin on pancreatic islets. In conclusion, the reduction in mRNA expression of the incretin receptors on rat pancreatic islets exposed to amylin, which could indicate an additional mechanism whereby amylin exert its deleterious effect on beta cells, reducing the insulinotropic effects of incretins in patients with T2DM was not confirm regarding GIPR and GLP1R protein expression at the time period studied. The increased mRNA expression of these receptors caused by amylin after 48 h of incubation could be a compensation mechanism against the toxic effects of amylin oligomers. The pro-apoptotic effect of amylin on human beta cells does not appear to be potentiated by lipotoxicity or by advanced glycation end products
305

Benfotiamina e Mito Q protegem ilhotas pancreáticas de rato em cultura dos efeitos pró-apoptóticos dos produtos finais de glicação avançada (AGEs) / Benfotiamine and Mito Q protect rat pancreatic islets in culture from pro-apoptotic effects of advanced glycation end products

Flavia Soares Louro Costal 13 March 2012 (has links)
A perda da função das células beta acelera a deterioração do controle metabólico em pessoas com diabetes tipo 2. Além da lipo- e da glicotoxicidade, os AGEs parecem contribuir para esse processo, promovendo a apoptose das ilhotas pancreáticas. Em outros tecidos, os AGEs interagem com seu receptor específico (RAGE), produzindo espécies reativas de oxigênio (ROS) e ativando o NF-kB. Para investigar o efeito temporal dos AGEs sobre a apoptose de ilhotas, bem como o potencial de compostos antioxidantes para diminuir danos causados pelos AGEs, ilhotas pancreáticas de ratos foram tratadas durante 24, 48, 72, 96 e 120 h com AGEs gerados a partir de co-incubação de albumina de soro bovino (BSA) com Dgliceraldeído (GAD, 5 mg/mL) ou tampão fostato (controle). A apoptose foi avaliada pela quantificação do DNA fragmentado (ELISA), atividade de caspase 3 e detecção da permeabilidade da membrana mitocondrial (MitoProbe JC-1). O estresse oxidativo foi avaliado pela detecção de espécies de oxigênio (Image-iT LIVE Green) e a atividade da NADPH oxidase foi mensurada pelo método de quimioluminescência da lucigenina. A expressão dos genes Bax, Bcl2 e Nfkb1 foi avaliada por reação em cadeia da polimerase quantitativa após transcrição reversa (RT-qPCR). Em um dos tempos em que foi detectado o aumento da apoptose, o efeito de dois compostos antioxidantes foi avaliado: benfotiamina (350 M), uma vitamina B1 lipossolúvel, e Mito Q (1 M), um derivado da ubiquinona com alvo seletivo para a mitocôndria. Em 24 e 48 h, os AGES promoveram um aumento do índice de apoptose em relação ao controle, concomitantemente com o aumento na expresssão do gene Bcl2 (gene anti-apoptótico) e uma redução na expressão do gene Nfkb1. Em contraste, após 72, 96 h e 120 h, os AGEs promoveram um aumento do índice de apoptose em comparação com a condição de controle, concomitantemente com uma diminuição na expressão do gene Bcl2 e um aumento na expressão do gene Nfkb1. Em 24 h, os AGEs promoveram uma diminuição do conteúdo de ROS nas ilhotas, enquanto que nos tempos de 48 e 72 h, os AGEs promoveram um efeito oposto. A benfotiamina e o Mito Q foram capazes de diminuir o índice de apoptose e o estresse oxidativo de ilhotas expostas aos AGEs por 72 h. Em conclusão, os AGEs exerceram um duplo efeito em cultura de ilhotas pancreáticas, sendo de proteção contra a apoptose após exposição curta, mas pró-apoptótica após exposição prolongada. O Mito Q e e a benfotiamina merecem ser adicionalmente estudados como drogas com o potencial de oferecer proteção às ilhotas pancreáticas em condições de hiperglicemia crônica / Loss of beta cell function hastens the deterioration of metabolic control in people with type 2 diabetes. Besides lipo- and glucotoxicity, AGEs seem to contribute to this process by promoting islet apoptosis. In other tissues, AGEs interact with their specific receptors (RAGE) and elicit reactive oxygen species (ROS) generation and NF-kB activation. In order to investigate the temporal effect of AGEs on islet apoptosis as well as the potential of antioxidant compounds to decrease islet damage caused by AGEs, rat pancreatic islets were treated for 24, 48, 72, 96 and 120 h with either AGEs generated from co-incubation of bovine serum albumin (BSA) with D-glyceraldehyde (GAD, 5 mg/mL) or phosphate-buffered saline (PBS, control). Apoptosis was evaluated by quantification of DNA fragmentation (ELISA), caspase-3 enzyme activity and detection of mitochondrial permeability transition (MitoProbe JC-1). Oxidative stress was evaluated by oxygen species detection (Image-iT LIVE Green) and the activity of NADPH oxidase was measured by the lucigenin-enhanced chemiluminescence method. The expression of the genes Bax, Bcl2 and Nfkb1 was evaluated by reverse transcription real-time quantitative polymerase chain reaction (RT-qPCR). In one of the time points at which increased apoptosis was detected, the effect of two antioxidant compounds was evaluated: benfotiamine (350 M), a liposoluble vitamin B1, and Mito Q (1 M), a derivative of ubiquinone targeted to mitochondria. In 24 and 48 h, AGEs elicited a significant decrease in the apoptosis rate in comparison to the control condition concomitantly with a significant increase in the RNA expression of the antiapoptotic gene Bcl2 and a significant decrease in the Nfkb1 RNA expression. In contrast, after 72 and 96 h, AGEs promoted a significant increase in the apoptosis rate in comparison to the control condition concomitantly with a significant decrease in Bcl2 RNA expression and a significant increase in Nfkb1 RNA expression. In 24 h, AGEs elicited a significant decrease in the islet content of ROS while after 48 and 72 h, AGEs promoted an opposite effect. Benfotiamine and Mito Q were able to decrease the apoptosis rate and the ROS content in islets exposed to AGEs for 72 h. In conclusion, AGEs exerted a dual effect in cultured pancreatic islets, being protective against apoptosis after short exposition but proapoptotic after prolonged exposition. Mito Q and benfotiamine deserve further evaluation as drugs that could offer islet protection in conditions of chronic hyperglycemia
306

Comparação dos efeitos do gangliosideo GM1 e do fator de crescimento neural (NGF) sobre a expressão de receptor de alta afinidade para NGF, TrkA e insulina em ilhotas pancreaticas isoladas de camundongos NOD (diabetico não obeso) / Comparison of the effect of ganglioside GM1 and the Nerve Growth Factor (NGF) on the expression of receiver of high affinity for NGF, TrkA and insulin in isolated pancreatic islets of NOD mice (non obese diabetic)

Domingos, Priscila Perez 29 February 2008 (has links)
Orientador: Ricardo de Lima Zollner / Tese (doutorado) - Universidade Estadual de Campinas, Faculdade de Ciencias Medicas / Made available in DSpace on 2018-08-11T22:15:47Z (GMT). No. of bitstreams: 1 Domingos_PriscilaPerez_D.pdf: 2379926 bytes, checksum: df7f068098f3454b58caf0a13e61f196 (MD5) Previous issue date: 2008 / Resumo: O camundongo não obeso diabético (NOD) é caracterizado por desenvolver naturalmente diabetes mellitus tipo 1 (DM-1) com similaridade ao diabetes mellitus tipo 1 em humanos. A manifestação espontânea do diabetes neste modelo animal é caracterizado por infiltração progressiva das ilhotas de Langerhans por células mononucleares linfócitos T (CD4+ e CD8+) e destruição das células ß pancreáticas produtoras de insulina. O fator de crescimento neural (NGF) e algumas citocinas estão associados a regeneração neural, além de atuarem sobre células do sistema imune. Em adição a estes efeitos, NGF age na liberação de insulina pelas células betas das ilhotas pancreáticas, tornando-se foco de interesse com relação as suas propriedades moduladoras no processo inflamatório na ilhota pancreática. O gangliosídeo GM1 liga-se ao receptor de alta afinidade (TrkA) do NGF-ß, mimetizando seus efeitos. No presente trabalho, avaliamos a ação modulatória de GM1 e NGF em cultura de ilhotas pancreáticas, provenientes de camundongos NOD. Foram avaliados por meio de RT-PCR a expressão gênica de NGF-ß, TrkA e insulina e, por ensaio imunoenzimático, a concentração de citocinas IL-1ß, IL-12, TNF-a, INF-y e insulina. Nossos resultados sugerem ação moduladora similar entre GM1 e NGF sobre as ilhotas de NOD não diabéticos e pré-diabéticos. NGF e GM1 aumentam a expressão gênica de NGF e TrkA e diminuem a expressão gênica de insulina em NOD não diabéticos e pré-diabéticos. Além disso, aumentam a liberação de insulina e diminui a de citocinas inflamatórias IL-1ß, IL-12, TNF-a, IFN-y que caracterizam a resposta Th1. / Abstract: The non-obese diabetic mice (NOD) lineage is characterized by developing type 1 diabetes mellitus (DM-1) naturally, bearing a similarity to DM-1 in human beings. The spontaneous manifestation of diabetes is characterized by gradual infiltration in pancreatic islets by mononuclear cells lymphocytes T (CD4+ and CD8+) and destruction of the ß-cells producers of insulin. One consequence of this effect, is the release of neurotrophins trying modulate the insulin release by the ß cells of pancreatic islets. Thus, the neurotrophins have been the focus of interest in the modulation of the inflammatory process in the pancreatic islets. The ganglioside GM1 binds to the high affinity receptor (TrkA) of the NGF-ß, enhancing its effect. In the present work, we evaluate the immune modulation properties of GM1 and NGF in culture of pancreatic islets from NOD mice. The gene expression of NGF-ß, TrkA and insulin for immune enzymatic assay, the concentration of cytokines IL 1ß, IL-12, TNF-a, IFN-y and insulin were evaluated by RT-PCR and ELISA. Our results suggest similar modulation action between GM1 and NGF on islets of NOD non-diabetic and pre-diabetic. GM1 and NGF action increases the gene expression of NGF and TrkA and the decrease of insulin in mice NOD non-diabetic and pre-diabetic. Moreover, GM1 and NGF increase the insulin release and decrease inflammatory cytokines that characterize the Th1 reply. / Doutorado / Ciencias Basicas / Doutor em Clínica Médica
307

Identification and characterization of the endoplasmic reticulum (ER)-stress pathways in pancreatic beta-cells / Identification et caractérisation des voies de signalisation du stress du réticulum endoplasmique dans la cellule bêta pancréatique

Pirot, Pierre 26 November 2007 (has links)
The endoplasmic reticulum (ER) is the organelle responsible for synthesis and folding of secreted and membranous protein and lipid biosynthesis. It also functions as one of the main cellular calcium stores. Pancreatic beta-cells evolved to produce and secrete insulin upon demand in order to regulate blood glucose homeostasis. In response to increases in serum glucose, insulin synthesis represents nearly 50% of the total protein biosynthesis by beta-cells. This poses an enormous burden on the ER, rendering beta-cells vulnerable to agents that perturb ER function. Alterations of ER homeostasis lead to accumulation of misfolded proteins and activation of an adaptive response named the unfolded protein response (UPR). The UPR is transduced via 3 ER transmembrane proteins, namely PERK, IRE-1 and ATF6. The signaling cascades activated downstream of these proteins: a) induce expression of ER resident chaperones and protein foldases. Increasing the protein folding capacity of the ER; b) attenuate general protein translations which avoids overloading the stressed ER with new proteins; c) upregulate ER-associated degradation (ERAD) genes, which decreases the unfolded protein load of the ER. In severe cases, failure by the UPR to solve the ER stress leads to apoptosis. The mechanisms linking ER stress to apoptosis are still poorly understood, but potential mediators include the transcription factors Chop and ATF3, pro-apoptotic members of the Bcl-2 familly, the caspase 12 and the kinase JNK. <p>Accumulating evidence suggest that ER stress contributes to beta-cell apoptosis in both type 1 and type 2 diabetes. Type 1 diabetes is characterized by a severe insulin deficiency resulting from chronic and progressive destruction of pancreatic beta-cells by the immune system. During this autoimmune assault, beta-cells are exposed to cytokines secreted by the immune cells infiltrating the pancreatic islets. Our group has previously shown that the pro-inflamatory cytokines interleukin-1beta (IL1-beta and interferon-gamma (IFN-gamma), via nitric oxide (NO) formation, downregulate expression and function of the ER Ca2+ pump SERCA2. This depletes beta-cell ER Ca2+ stores, leading to ER stress and apoptosis. Of note, IL1-beta alone triggers ER stress but does not induce beta-cell death, while IFN-gamma neither causes ER stress nor induces beta-cell death. Together, these cytokines cause beta-cell apoptosis but the mechanisms behind this synergistic effect were unknown.<p>Type 2 diabetes is characterized by both peripheral resistance to insulin, usually as a result of obesity, and deficient insulin secretion secondary to beta cell failure. Obese patients have high levels of circulating free fatty acids (FFA) and several studies have shown that the FFA palmitate induces ER stress and beta-cell apoptosis.<p>In the present work we initially established an experimental model to specifically activate the ER stress response in pancreatic beta-cells. For this purpose, insulinoma cells (INS-1E) or primary rat beta-cells were exposed to the reversible chemical SERCA pump blocker cyclopiazonic acid (CPA). Dose-response and time course experiments determined the best conditions to induce a marked ER stress without excessive cell death (<25%).<p>The first goal of the work was to understand the synergistic effects of IL1-beta and IFN-gamma leading to pancreatic beta-cell apoptosis. Our group previously observed, by microarray analysis of primary beta-cells, that IFN-gamma down-regulates mRNAs encoding for some ER chaperones. Against this background, our hypothesis was that IFN-gamma aggravates beta-cell ER stress by decreasing the ability of these cells to mount an adequate UPR. To test this hypothesis, we investigated whether IFN-gamma pre-treatment augments CPA-induced ER stress and beta cell death. The results obtained indicated that IFN-gamma pre-treatment potentiates CPA-induced apoptosis in INS-1E and primary beta-cells. This effect was specific for IFN-gamma since neither IL1-beta nor a low dose CPA pre-treatment potentiated CPA-induced apoptosis in INS-1E cells. These effects of IFN-gamma were mediated via the down regulation of genes involved in beta cell defense against ER stress, including the ER chaperones BiP, Orp150 and Grp94 as well as Sec61, a component of the ERAD pathway. This had functional consequences as evidenced by a decreased basal and CPA-induced activity of a reporter construct for the unfolded protein response element (UPRE) and augmented expression of the pro-apoptotic transcription factor Chop. <p>We next investigated the molecular regulation of the Chop gene in INS-1E cells in response to several pro-apoptotic and ER stress inducing agents, namely cytokines (IL1-beta and IFN-gamma), palmitate, or CPA. Detailed mutagenesis studies of the Chop promoter showed differential regulation of Chop transcription by these compounds. While cytokines (via NO production)- and palmitate-induced Chop expression was mediated via a C/EBP-ATF composite and AP-1 binding sites, CPA induction required the C/EBP-ATF site and the ER stress response element (ERSE). Cytokines, palmitate and CPA induced ATF4 protein expression and further binding to the C/EBP-ATF composite site, as shown by Western blot and EMSA experiments. There was also formation of distinct AP-1 dimers and binding to the AP-1 site after exposure to cytokines or palmitate. <p>\ / Doctorat en Sciences biomédicales et pharmaceutiques / info:eu-repo/semantics/nonPublished
308

Molecular pathways underlying beta-cell loss in vitro models of type 2 diabetes mellitus

Kharroubi, Ilham January 2006 (has links)
Doctorat en Sciences médicales / info:eu-repo/semantics/nonPublished
309

Proliferação e disfunção da célula beta pancreática em modelo animal de Diabetes Melito tipo 2. Envolvimento da via de sinalização WNT/Beta-Catenina / Pancreatic beta cell proliferation and dysfunction in animal model of type 2 Diabetes Mellitus. Involvement of the WNT/Beta-catenin signaling pathway

Oliveira, Ricardo Beltrame de 18 August 2018 (has links)
Orientador: Carla Beatriz Collares Buzato / Dissertação (mestrado) - Universidade Estadual de Campinas, Instituto de Biologia / Made available in DSpace on 2018-08-18T15:01:37Z (GMT). No. of bitstreams: 1 Oliveira_RicardoBeltramede_M.pdf: 4343326 bytes, checksum: c45de34d36b3510fa7bea078864506dc (MD5) Previous issue date: 2011 / Resumo: Tem havido um grande interesse na determinação das vias envolvidas na proliferação e função/disfunção da célula beta e a aplicação deste conhecimento em terapias moleculares e celulares da diabetes. A patogênese da diabetes melito tipo 2 (T2DM) é complexa, mas frequentemente está associada com obesidade e distúrbios do metabolismo de lipídios (hipercolesterolemia e hipertrigliceridemia). A T2DM envolve o desenvolvimento de um quadro de resistência periférica à insulina parcialmente compensada por hiperinsulinemia e hiperplasia da célula beta pancreática, resultando em intolerância à glicose e hiperglicemia. Os mecanismos interligando os estados de obesidade/hipercolesterolemia e resistência à insulina ao fenômeno da hiperplasia da célula beta não são completamente conhecidos. A presente dissertação teve como objetivos: 1) caracterizar um modelo animal adequado para se estudar a proliferação e disfunção da célula beta pancreática, e 2) avaliar, no pâncreas endócrino desses animais, a possível ativação da via de sinalização Wnt/beta-catenina, conhecida por estar envolvida no processo de proliferação celular em outros tecidos/órgãos. Para tal, foram empregados camundongos C57BL/6, wild-type (WT) e knockout para receptor de lipoproteína LDL (LDLr-/-), os quais foram submetidos à dieta hiperlipídica (HF) por 60 dias. Após a dieta HF, os animais WT tornaram-se obesos e hipercolesterolêmicos, bem como moderadamente hiperglicêmicos, hiperinsulinêmicos, intolerantes à glicose e resistentes à insulina, caracterizando-os como pré-diabéticos. Além disso, os animais alimentados com dieta HF apresentaram uma diminuição significativa na resposta secretora das células beta à glicose. De modo geral, os animais LDLr-/- apresentaram uma susceptibilidade relativamente mais alta à dieta HF, sugerida pela acentuada hipercolesterolemia, intolerância à glicose, e reduzida secreção de insulina estimulada por glicose observadas nestes animais. No entanto, a dieta HF induziu, de forma semelhante em animais WT e LDLr-/-, uma diminuição significativa no conteúdo celular de Cx36, uma proteína associada à junção comunicante e um marcador de diferenciação terminal da célula beta. Ambos os grupos WT e LDLr-/- alimentados com dieta HF mostraram aumento na proliferação de células beta, como avaliada pela imunomarcação das ilhotas para a proteína Ki67, mas apenas os animais WT exibiram alterações morfométricas indicativas de hiperplasia do pâncreas endócrino, tais como aumento na massa total de ilhotas e de células beta. Uma vez estabelecido que camundongos WT alimentados com dieta HF por 60 dias consistiam em um modelo adequado para a segunda etapa deste estudo, fomos investigar a possível ativação da via Wnt/beta-catenina nas ilhotas pancreáticas desses animais, avaliando-se a distribuição e expressão celular das proteínas beta-catenina total, beta-catenina ativada, c-Myc e ciclina D. A análise por imunofluorescência para beta-catenina não mostrou acúmulo citoplasmático ou translocação para o núcleo desta proteína em ilhotas pancreáticas, que poderia indicar ativação da via Wnt/beta-catenina no nosso modelo de hiperplasia do pâncreas endócrino. No entanto, a análise por Western Blot revelou um aumento significativo na expressão de beta-catenina ativada e ciclina D em ilhotas de animais alimentados com dieta HF em relação ao grupo controle. Concluindo, a dieta HF por 60 dias induz alterações metabólicas típicas da pré-diabetes em animais WT e LDLr-/-. O estado de pré-diabetes está associado a uma diminuição da expressão de Cx36 nas células beta pancreáticas, sugerindo um possível papel da comunicação intercelular mediada pelas junções comunicantes na patogênese da T2DM. A maior susceptibilidade metabólica à dieta HF apresentada por camundongos LDLr-/-, em relação aos WT, pode ser explicada pela maior deficiência na secreção de insulina em resposta à glicose e ausência de hiperplasia compensatória do pâncreas endócrino. Ainda, a análise preliminar de expressão protéica de algumas proteínas da via Wnt/beta-catenina sugere que esta via parece estar ativada durante o processo de hiperplasia do pâncreas endócrino observada no nosso modelo animal / Abstract: The pathogenesis of type 2 diabetes mellitus (T2DM) is often associated with obesity and dyslipidemia (hypercholesterolemia and hypertriglyceridemia). T2DM involves intolerance to glucose and insulin resistance partially compensated by hyperinsulinemia and pancreatic beta cell hyperplasia. The mechanisms linking obesity/hypercholesterolemia and insulin resistance to beta cell hyperplasia are not fully known. The Wnt/beta-catenin signaling pathway has been reported to be involved in cell growth and differentiation in several tissues/organs but its role in endocrine pancreas development and function is still unclear. This work aimed at: 1) establishing an appropriate animal model of T2DM to study pancreatic beta cell proliferation and dysfunction and, 2) investigating a putative involvement of the Wnt/beta-catenin signaling pathway in the beta cell hyperplasia in this model. To this end, we employed C57BL/6 wild-type (WT) and LDL lipoprotein receptor knockout (LDLr-/-) mice, fed a high fat (HF) diet for 60 days. After feeding a HF diet, WT mice became obese, hypercholesterolemic and moderately hyperglycemic, hyperinsulinemic, glucose intolerant and insulin resistant, characterizing them as pre-diabetics. Moreover, animals fed a HF diet showed a significant decrease in beta-cell secretory response to glucose. In general, LDLr-/- animals showed a relatively higher susceptibility to HF diet, as suggested by a marked hypercholesterolemia, glucose intolerance and reduced insulin secretion stimulated by glucose observed in these animals as compared to the control ones. However, HF diet induced similarly in both WT and LDLr-/- mice a significant decrease in cellular content of Cx36, a gap junctional protein and marker of terminally differentiated beta cell. Both WT and LDLr-/- fed a HF diet showed increased proliferation of beta cells, as assessed by Ki67 immunostaining, but only WT mice exhibited morphometric changes indicative of endocrine pancreas hyperplasia, such as increased total islet and beta cell masses. After we investigated a possible activation of Wnt/beta-catenin signaling pathway in these hyperplasic pancreatic islets of WT animals fed a HF diet. This was done by assessing the distribution and cellular protein expression of some proteins associated to this pathway (i.e., total and activated beta-catenin, c-Myc and cyclin D) in islets of our animal model. Beta-catenin immunofluorescence showed no cytoplasmic accumulation or translocation into the nucleus of beta cells in HF-fed mice. However, immunoblotting revealed a significant increase of unphosphorylated beta-catenin (activated) and cyclin D expression in islets of HF diet-fed animals when compared to its control group. In conclusion, a HF diet for 60d induced pre-diabetes state in both WT and LDLr-/- mice. The pre-diabetes state is associated with a decreased expression of Cx36 in pancreatic beta cells, suggesting a possible role of intercellular communication mediated by gap junctions in the pathogenesis of T2DM. The relatively high metabolic susceptibility to the HF diet showed by LDLr-/- mice, as compared to WT, may be explained by a marked impairment of glucosestimulated insulin secretion and a lack of compensatory hyperplasia of the endocrine pancreas. In addition, the protein expression analysis suggests that the Wnt/beta-catenin pathway may be activated during the islet hyperplasia process in our animal model / Mestrado / Histologia / Mestre em Biologia Celular e Estrutural
310

Étude de la fonction du récepteur aux acides gras GPR120/FFAR4 dans la régulation de l’homéostasie du glucose

Guillaume, Arthur 05 1900 (has links)
Le diabète de type 2 (DT2) résulte de l’incapacité des cellules β sécrétrices d’insuline à compenser la résistance à l’insuline qui s’installe chez les patients obèses. Un traitement potentiel viserait donc à augmenter la sécrétion d’insuline. Dans ce sens, les récepteurs aux acides gras GPR120 et GPR40 potentialisent la sécrétion d’insuline. Cependant, la signalisation GPR120 dans les îlots est méconnue. L’activation de GPR120 diminue la sécrétion de somatostatine (SST), un inhibiteur de la sécrétion d’insuline, par les cellules δ. Ces deux récepteurs régulent l’homéostasie du glucose et sont donc possiblement complémentaires. Nos objectifs étaient d’étudier la signalisation GPR120 dans les îlots pancréatiques, ainsi que la complémentarité des récepteurs GPR120 et GPR40 dans le contrôle de l’homéostasie glucidique. À l’aide d’îlots isolés de souris n’exprimant pas GPR120, constitutivement ou uniquement dans les cellules δ, nous avons étudié le rôle de GPR120 dans les sécrétions d’insuline, glucagon et de SST. Nous avons ensuite étudié des souris n’exprimant pas GPR40, GPR120 ou les deux, sous une diète riche en gras pendant 12 semaines pour étudier la complémentarité des deux récepteurs. L’activation de GPR120 diminue la sécrétion de SST et stimule les sécrétions d’insuline et de glucagon dans les îlots isolés. Cet effet est aboli par la délétion de GPR120 dans les cellules δ in vitro, et la double délétion de GPR120 et GPR40 ne révèle pas d’action complémentaire dans l’homéostasie glucidique. Ces résultats suggèrent que la signalisation GPR120 dans les cellules δ est responsable de l’amélioration de la fonction des îlots. Une meilleure compréhension du rôle joué par GPR120 dans la fonction des îlots et l’homéostasie du glucose est cruciale et pourrait permettre le développement de nouvelles options thérapeutiques dans le traitement du diabète. / In obese patients, type 2 diabetes stems from the failure of the insulin-secreting beta cells to compensate for insulin resistance. Increasing insulin secretion is therefore a viable treatment strategy. In this regard, G protein-coupled receptors (GPCR) are proven therapeutic targets. Activation of the GPCR for long-chain saturated and unsaturated fatty acid GPR40 and GPR120 increase insulin secretion in response to glucose. However, exactly how GPR120 potentiates insulin secretion is unknown. GPR120 and GPR40 both regulate glucose homeostasis and therefore could act in a complementary manner. We aimed to decipher GPR120 signalling in the pancreatic islets and study the complementary roles of GPR120 and GPR40 in maintaining glucose homeostasis. To this aim, we first measured insulin, glucagon and somatostatin secretion following GPR120 activation in isolated islets from mice with a global or somatostatin-cell-specific knock-out of GPR120. Then we studied glucose metabolism in mice with global deletion of GPR120, GPR40 or both, under a high fat diet for 12 weeks. We observed increased insulin and glucagon secretions mirrored by a decreased in somatostatin release following GPR120 activation in isolated islets, an effect abolished by a global or δ-specific deletion of GPR120. A double deletion of GPR120 and GPR40 did not have more impact on glucose metabolism or beta-cell function compared to a simple deletion of either receptor. A better understanding of the GPR120 role in islet function is crucial and could lead to the discovery of new therapeutic options.

Page generated in 0.072 seconds