• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 2
  • 2
  • Tagged with
  • 4
  • 4
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 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.
1

Studies On The Mechanisms Involved In Thymic Atrophy During Salmonella Enterica Serovar Typhimurium Infection

Deobagkar-Lele, Mukta 07 1900 (has links) (PDF)
T lymphocytes are an essential component of the adaptive immune response and are highly versatile in function. Each T cell has a unique T cell receptor that can recognize an antigenic peptide in the context of the major his to compatibility complex (MHC) encoded molecules, thus offering a high degree of specificity to the immune response. T cells play a central role in the development of an effective host immune response and the quantitative and qualitative regulation of the T cell response is critical. T cells develop in the thymus, an important primary immune organ, where immature thymocytes undergo differentiation and maturation. Through the process of thymic differentiation, immature cluster of differentiation (CD)4-CD8- thymocytes progress to a CD4+CD8+ stage and are subjected to positive and negative selection to give rise to MHC restricted, single positive CD4+ or CD8+ naive T cells that emigrate from the thymus and populate the peripheral lymphocyte pool. Thymic atrophy is well known to occur naturally during the process of aging with thymocyte depletion and reduced thymic output. Along with age associated changes leading to atrophy, the thymus is exquisitely sensitive to starvation and several stresses. In addition, thymic atrophy is a characteristic feature during several viral, bacterial and parasitic infections. Egress of immature thymocytes, loss of thymic populations due to sensitivity to glucocorticoids and cytokine modulation, etc. have been variously proposed to be involved in this process. However there is limited understanding on the numerous mechanisms involved and the crosstalk between these diverse pathways. In this study, a model for thymic atrophy during acute Salmonella enterica serovar Typhimurium (S. typhimurium) infection was developed. S. typhimurium is a Gram negative bacterium that resides and grows in intracellular compartments within host cells. It causes gastroenteritis in humans but leads to typhoid like disease in mice, similar to that caused by S. typhi in humans. Initially, it was established that acute infection of C57BL/6 mice with 108 CFU S. typhimurium, via the oral, i.e. the physiological, route of infection leads to extensive depletion (8-10 fold) of thymocytes in an infection-dependent manner. Infected mice had higher CFU burden in the Peyer’s patches, spleen, liver, and mesenteric lymph node (MLN) as compared to the thymus. The thymic atrophy was dependent upon the infection caused by live S. typhimurium since oral feeding of mice even with higher doses (1010 CFU) of heat-killed bacteria did not lead to thymic atrophy. The susceptible populations in the thymus were identified by staining for expression of CD4 and CD8 on cell surface using specific monoclonal antibodies tagged to fluorophores, e.g. Fluorescein isothiocyanate (FITC) and phycoerythrin (PE), respectively. The double labelled samples were analyzed by flow cytometry. Interestingly, significant death of CD4+CD8+, the major population of thymocytes, but not single positive thymocytes or peripheral lymphocytes (MLN and spleen cells), was observed at later stages during infection. To gain greater understanding of the processes involved, the mechanisms leading to thymic atrophy were investigated. To this purpose, small molecule inhibitors and mice lacking key molecules important for the immune response were utilized. Also, various assays to assess death of thymocytes, including analysis of death markers such as Annexin V based detection of membrane flipping and caspase activation were performed. I. The extrinsic death pathway involving Fas/FasL interactions is a major death pathway. Therefore, the expression and functional role of the components of the pathway in this model of thymocyte death was investigated. It was observed that thymocytes from infected mice expressed more Fas and Fas ligand (FasL) on their surface than cells from uninfected mice. To address the role of the death receptor, Fas, infection studies were performed with lpr mice that lack functional Fas expression. The depletion of CD4+CD8+ thymocytes in lpr mice was comparable to that in C57BL/6 mice indicating that it was independent of the Fas pathway. However, extensive loss of mitochondrial membrane potential was observed upon analysis with mitochondrial potential specific dyes MitoTracker Red and DiOC6. Most likely, the intrinsic death pathway involving mitochondrial depolarization is involved in this model of thymic atrophy. II. Since thymocytes are known to be sensitive to glucocorticoids both in vitro and in vivo, the involvement of the same in this model of thymic atrophy was assessed. The amounts of cortisol, a glucocorticoid, as detected by ELISA, were elevated during infection. To investigate the functional implication of the increase in cortisol, studies were performed using RU486, a glucocorticoid receptor antagonist. RU486 did not modulate cortisol amounts and treatment of mice with RU486 did not affect CFU burden or survival of mice. However there was a moderate rescue in the number of viable CD4+CD8+ thymocytes, with only a 3-4 fold drop as compared to the 8-10 fold drop in vehicle treated infected mice. III. As glucocorticoids appeared to play a partial role in this model, it was reasonable to assume that other pathways were also involved in the thymic atrophy. The quantitative and qualitative modulation of the cytokine milieu has a profound effect upon the thymus. In fact, inflammatory cytokines, Tnfα and Ifnγ, increased upon infection. In order to study the role of Ifnγ mediated inflammatory responses in this model, infection studies with Ifnγ-/- mice were performed. Ifnγ-/- mice had higher CFU and lower survival; however the drop in thymocyte numbers was 3-4 fold as compared to the 8-10 fold drop in the infected C57BL/6 mice, again indicating a partial involvement of the Ifnγ mediated pathways. In order to study the interactions, if any, between the two pathways mentioned above, corticosteroid signaling was blocked in the Ifnγ-/- mice with RU486. Upon infection, the number of CD4+CD8+ thymocytes was significantly higher in Ifnγ-/- mice treated with RU486 (~1.5 fold drop in viable thymocyte numbers) along with lower caspase 3 activity and mitochondrial damage. Importantly, cortisol amounts in infected Ifnγ-/- mice were comparable to those in infected C57BL/6 mice and the administration of RU486 did not modulate Tnfα and Ifnγ cytokine amounts in sera. Thus, the glucocorticoid and Ifnγ mediated pathways are parallel but synergize in an additive manner to induce death of CD4+CD8+ thymocytes during S. typhimurium nfection. IV. Although thymic atrophy is known to occur, a detailed characterization of cell surface changes in thymocyte populations has not been performed. To investigate this aspect, thymocytes and MLN cells from uninfected and infected animals were stained for cell surface expression of CD3, CD4, CD5, CD8, CD24, CD25, CD44, CD69, MHC I and MHC II. This analysis was initially performed by studying the changes in expression of these molecules within the total thymocyte and MLN populations. Although there was no change in the expression of CD25 and MHC II in the total thymocyte population upon infection, CD24 expression reduced, whereas, the expression of CD3, CD5, CD44, CD69 and MHC I increased. Notably, changes in the frequency of expression of CD3, CD69 and MHC I were observed before the development of extensive thymic atrophy. The depletion of majority of the CD4+CD8+ thymocytes enriches the mature CD4+ or CD8+ thymocyte population This was corroborated with the observation that, upon in vitro stimulation with PMA and Ionomycin (pharmacological agents used to activate T cells) the residual thymocytes from infected mice produced more IL2 compared to thymocytes from uninfected mice. Subsequently, cells were stained with anti-CD4-FITC, anti-CD8-PE and a third biotinylated antibody, which was detected by a streptavidin-APC conjugate, against one of the remaining six markers. This three colour analysis made it possible to determine the changes in the expression of the third marker in each of the CD4-CD8-, CD4+CD8+, CD4+ and CD8+ populations upon infection. Distinct differences were observed in the phenotypes of uninfected and infected CD4+CD8+ thymocytes and the latter were CD3high, CD5high, CD24low, CD69high and MHC Ihigh indicating that the surviving population had a possibly more mature phenotype. Also, the changes in the phenotypes of the thymocyte populations were dependent upon the extent of thymic atrophy as indicated by time course and CFU studies with C57BL/6 and BALB/c mice respectively. Finally, the roles of glucocorticoids, Ifnγ and Nos2 in modulation of expression of these markers during infection were addressed. Interestingly, the expression of CD3, CD24 and MHC class I significantly correlated with increase in the number of surviving thymocytes upon inhibition of glucocorticoids signaling and in Ifnγ-/- mice. The implications of these changes in the thymocyte surface phenotype during thymic atrophy are discussed. V. Finally, the roles of downstream signalling molecules in S. typhimurium induced thymic atrophy were studied. Although the MAP kinase family members, Erk, Jnk and p38 have been implicated to play a role in the positive and/or negative selection of thymocytes during development, their role in infection induced thymocyte depletion has not been studied. Interestingly, the amounts of Jnk and pJnk, but not p38, increased in thymocytes upon infection. Importantly, pJnk amounts increased predominantly in CD3-/low thymocytes during infection. Furthermore, inhibition of Jnk signalling, using a specific inhibitor SP600125, lead to an increase in survival of CD4+CD8+ thymocytes during infection due to multiple reasons: lowering of cortisol, Tnfα and Ifnγ amounts, and better maintenance of thymic architecture. Thus, inhibition of Jnk mediated signaling protected CD4+CD8+ and CD3-/low thymocytes from death during S. typhimurium infection. Overall, the main conclusions of this study are as follows: First, extensive analysis of the surface phenotype of cells during thymic atrophy throws light on the sensitive and resistant thymocyte populations, thus offering a potential predictive marker profile. Second, glucocorticoids, Ifnγ and, importantly, Jnk mediated signaling play functional roles in the death of immature CD4+CD8+ thymocytes during S. typhimurium infection. The mechanistic details uncovered in this study may be important in designing effective strategies for reducing thymic atrophy during other infections. In fact, enhancement of thymic output may lead to greater numbers and diversity of thymic T cell emigrants in the periphery which is likely to enhance host responses during infections.
2

Leptin Regulation of Thymopoiesis During Endotoxin-Induced Acute Thymic Atrophy

Gruver, Amanda Louise January 2009 (has links)
<p>Thymus atrophy is highly inducible by stress and prolonged thymus atrophy can contribute to T cell deficiency or inhibit immune recovery after acute peripheral T cell depletion. Little is known regarding the mechanisms driving thymic involution or thymic reconstitution after acute stress. Leptin deficiency in mice results in chronic thymic atrophy, suppressed cell-mediated immunity, and decreased numbers of total lymphocytes, suggesting a role for leptin in regulating thymopoiesis and overall immune homeostasis. Exogenous leptin administration during stress has been shown to protect against thymic damage, yet the mechanisms governing these thymostimulatory effects are currently undefined. Studies herein define the impact of endotoxin-induced thymic damage in the stromal and lymphoid compartment of the thymus and systemic glucocorticoid and cytokine responses in the animal. We report here the novel finding that leptin receptor expression is restricted to medullary thymic epithelial cells in the normal thymus. Using a model of endotoxin-induced acute thymic involution and recovery, we have demonstrated a role for the metabolic hormone leptin in protection of medullary thymic epithelial cells from acute endotoxin-induced damage. We also demonstrated that systemic leptin treatment decreased endotoxin-induced apoptosis of double positive thymocytes and promoted proliferation of double negative thymocytes in vivo through a leptin receptor isoform b-specific mechanism. Leptin treatment increased thymic expression of IL-7, an important soluble thymocyte growth factor produced by medullary thymic epithelial cells. We also found leptin to inhibit systemic glucocorticoid and pro-inflammatory cytokine responses. Using leptin-deficient and leptin receptor-deficient mice in our stress model, we found that endotoxin-induced thymic atrophy was exacerbated in the absence of leptin, despite an inability to mount a proper pro-inflammatory cytokine response. Together, these data support a model in which leptin can function to protect the thymus gland from stress-induced acute damage in part by reduction of systemic corticosteroid and pro-inflammatory cytokine responses, and intrathymically through a mechanism orchestrated by medullary thymic epithelial cells and their soluble mediators (e.g. IL-7). Taken together, these studies suggest a physiological role for leptin signaling in the thymus for maintaining healthy thymic epithelium and promoting thymopoiesis, which is revealed when thymus homeostasis is perturbed by stress.</p> / Dissertation
3

Aterações no microambiente timico frente a diferentes agentes indutores de atrofia / Thymic microenvironmental alterations in different kind of thymus atrophy

Gameiro, Jacy 13 August 2018 (has links)
Orientadores: Liana Verinaud, Wilson Savino / Tese (doutorado) - Universidade Estadual de Campinas, Instituto de Biologia / Made available in DSpace on 2018-08-13T05:50:49Z (GMT). No. of bitstreams: 1 Gameiro_Jacy_D.pdf: 7674475 bytes, checksum: 85104c8f2befe30630bf49436c0fb352 (MD5) Previous issue date: 2009 / Resumo: O timo é o órgão linfóide primário responsável pelo amadurecimento dos linfócitos T. O processo de maturação é dependente da integridade do microambiente e da migração coordenada por elementos de matriz e quimiocinas dos precursores hematopoiéticos nos distintos nichos tímicos. Entretanto, a literatura mostra que profundas alterações no microambiente tímico, e conseqüentemente no desenvolvimento e amadurecimento dos linfócitos T, podem ocorrer como resultado de algumas patologias. No presente trabalho, analisamos a indução de atrofia e as alterações no microambiente tímico frente a três diferentes agentes indutores de atrofia, sendo dois agentes patogênicos, P.brasiliensis e P.berghei e uma doença metabólica, a diabetes induzida por aloxana. Nos três modelos estudados foi observada a atrofia do órgão com diferente intensidade nas alterações de componentes do microambiente tímico. Importantes moléculas associadas com a migração intratímica mostraram-se alteradas com modificações significativas nos elementos de matriz e nas quimiocinas bem como na expressão dos seus respectivos receptores. Ainda, a migração ex vivo também se mostrou alterada na atrofia induzida pelos agentes patogênicos, mas não no modelo de atrofia induzido por aloxana, sugerindo que as alterações desencadeadas são específicas de cada condição patológica analisada. As modificações nos elementos estudados sugerem alterações no padrão de migração intratímica e na exportação de timócitos, com comprometimento da função tímica, levando a maturação desequilibrada dos timócitos, com conseqüências para a resposta imune periférica. / Abstract: Thymus is the primary lymphoid organ responsible for differentiation of Tlymphocytes. This process is dependent of thymic microenvironment integrity and coordinated migration of hematopoietic precursors by chemokines and extracellular matrix elements. However, the literature shows that deep alterations in thymus microenvironment with modifications in thymocyte development may occur such as result of some pathological disorders. In this study we analyzed the atrophy induction and alterations in thymic microenvironment in three different models of thymus atrophy. We studied two models of infectious diseases, P.brasiliensis and P.berghei, and a metabolic disorder, alloxan induced diabetes. We have observed thymus atrophy in all models with different alterations levels in thymus microenvironment elements. Important molecules associated with intrathymic migration were been altered with significant modifications in extracellular matrix elements, chemokines and their specific receptors. Besides, ex vivo migration was altered in thymus atrophy induced by pathogenic agents, but no alterations were observed in diabetic mice suggesting that different pathological conditions studied, leads to singular alterations in thymus compartment. The modifications in thymic molecules observed in our models, suggest impaired thymus functionality and alterations in thymocyte migration patterns These alterations can lead T cell maturation imbalance with consequences in Tlymphocyte immune response. / Doutorado / Imunologia / Doutor em Genetica e Biologia Molecular
4

Comprometimento timico em tres diferentes modelos experimentais de atrofia timica : alterações celulares e de citocinas / Thymic cells and cytokines alterations in different kind of thymus atrophy

Loyola, Patricia Resende Alo Nagib 13 August 2018 (has links)
Orientador: Liana Verinaud, Dea Maria Serra Villa-Verde / Tese (doutorado) - Universidade Estadual de Campinas, Instituto de Biologia / Made available in DSpace on 2018-08-13T21:07:32Z (GMT). No. of bitstreams: 1 Loyola_PatriciaResendeAloNagib_D.pdf: 6019241 bytes, checksum: 171494f158eff0d6b54b029e9583a35c (MD5) Previous issue date: 2009 / Resumo: O timo é o órgão linfóide primário onde os precursores de células T, chamados de timócitos, sofrem processos de diferenciação, seleção e proliferação. Todos estes processos são seqüencialmente dependentes de distintos microambientes no interior do órgão delimitados morfológica e fenotipicamente. Por isso a manutenção do microambiente é essencial para a funcionalidade do órgão. Recentemente, o timo tem sido visto como um órgão alvo de diversas infecções e patologias. No presente trabalho, foi avaliado o comprometimento do timo em três diferentes modelos experimentais capazes de induzir sua atrofia, a saber, infecção experimental pelo fungo P. brasiliensis, infecção experimental por Plasmodium berghei e diabetes induzida quimicamente. Foram avaliadas as subpopulações celulares tímicas, a expressão de citocinas essenciais à maturação dos timócitos e o possível papel do hormônio leptina no estabelecimento e manutenção da atrofia tímica. Sucintamente, os resultados demonstraram que nos três modelos, houve diminuição de celularidade com alterações significativas na freqüência dos subtipos de timócitos, principalmente queda de DP, e menor número de TNCs/animal . No modelo de diabetes além destas alterações, foi detectado a queda dos níveis de leptina, aparentemente, devido á queda da insulina. No modelo de PCM, não houve alteração nos níveis de leptina, entretanto, houve queda na expressão gênica de IL-7 e TGF-ß, fatores timo-estimulantes e inibição da capacidade migratória. Em animais infectados pelo P. berghei, houve queda de leptina e queda na expressão dos genes específicos para IL-7 e TGF-ß, além de aumento na atividade de migração Mediante os dados obtidos em cada modelo, sugere-se que no caso do modelo de PCM a atrofia de grau leve que foi observada deve-se à perda de células por diminuição da expressão das citocinas timo-estimulantes. Em malária acredita-se que a intensa atrofia ocorra por perda da região cortical devido, à queda na expressão das citocinas IL-7, TGF-ß e nos níveis séricos de leptina. Na diabetes induzida, o grau intermediário de atrofia pode ser correlacionado apenas à queda dos níveis séricos de leptina. / Abstract: The thymus is the primary lymphoid organ in which T cell precursors, called thymocytes, undergo processes of differentiation, selection, and proliferation. All these processes are sequentially dependent on distinct thymic microenvironments, which in turn, are delimited morphologically and phenotypically. For these reasons, the maintenance of this microenvironment is crucial for thymus functionality. Recently, the thymus has been seen an organ targeted by several infections and pathologies. In this study, we evaluated thymus compromise under three experimental models capable of inducing atrophy, as follows: (1) experimental infection by the fungus Paracoccidioides brasiliensis (PCM), (2) experimental infection by Plasmodium berghei, and (3) chemically-induced diabetes. We evaluated subpopulations of thymic cells, the expression of cytokines that are essential to thymocyte maturation, and the possible role of the leptin hormone in the establishment of thymic atrophy. Briefly, the results showed that in all three models there was a decrease of cellularity along with significant alterations in the frequency of each thymocyte subtype, especially a decrease in DP, and lower numbers of TNCs per animal. In the diabetes model, besides these alterations, we detected a decrease in leptin levels, apparently related to the insulin drop. In the PCM model, there were no alterations in leptin levels. However, we observed a drop in the expression of IL-7 and TGF- ß, both thymus-stimulant factors, as well as an inhibition of migratory ability. In P. berghei-infected animals there was a drop in leptin as well as in the expression of IL-7 and TGF- ß genes; besides an increase in migration activity. According to the data obtained within each model, we suggest that for the PCM model, the low degree of observed atrophy is a consequence of the loss of cells due to a drop in thymus-stimulant cytokine expression. In malaria, the intense atrophy happens due to the loss of the cortex region, generated by the drop in the expression of IL-7 cytokines, TGF- ß, and seric levels of leptin. As for the induced diabetes, the intermediate degree of atrophy can be correlated only to the drop in seric levels of leptin. / Doutorado / Imunologia / Doutor em Genetica e Biologia Molecular

Page generated in 0.051 seconds