• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 10
  • 8
  • 2
  • 2
  • Tagged with
  • 24
  • 24
  • 24
  • 8
  • 8
  • 7
  • 7
  • 7
  • 6
  • 6
  • 6
  • 5
  • 4
  • 4
  • 4
  • 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.
21

Investigação da resposta imunológica antitumoral induzida por células B16F10 tratadas pela combinação p19Arf e interferon-beta em um modelo de vacinação profilático para melanoma murino / Investigation of the antitumor immune response induced by B16F10 cells treated with the p19Arf and Interferon-beta combination in a murine prophylatic model of melanoma vaccine

Ruan Felipe Vieira Medrano 25 April 2013 (has links)
Dados recentes do nosso laboratório demonstram que somente a co-transdução, não a tradução individual, com vetores adenovirais portadores de Interferon-beta (IFN?) (citocina imuno modulatória) e p19Arf (parceira funcional da proteína supressora de tumor p53) resulta na morte celular massiva do melanoma murino B16F10. A capacidade desse tratamento combinado de induzir uma resposta imune antitumoral ainda não foi avaliada. Dessa maneira, o objetivo do presente trabalho foi investigar se células B16F0 tratadas por essa combinação são capazes de induzir uma resposta imune antitumoral em um modelo de vacinação profilático de melanoma. Para isso, essas células foram co-transduzidas com os vetores AdPGp19 e AdPGIFN? e 48 horas depois, inoculadas como agente vacinal no flanco esquerdo (sítio da vacina) de camundongos C57BL/6 imunocompetentes. Sete dias após a última vacinação, esses animais foram desafiados com células B16F10 naïve no flanco direito (sítio do desafio). A progressão tumoral do desafio foi significativamente reduzida, mesmo quando o desafio tumoral foi feito 73 dias após da vacinação. Porém, como os animais imunizados desenvolveram tumores no sítio da vacina, condições para o uso dessas células tratadas foram avaliadas, revelando que: o número de células e de aplicações usadas durante a vacinação tem influência no aparecimento desse tumores, e que apenas com o tratamento combinado os camundongos permanecem livres de tumor. A influência do sistema imune para este resultado foi revelada após protocolo de imunussupressão. Em seguida, o papel da p19Arf e do IFN? na proteção antitumoral da combinação foi estudado. In vitro, os efeitos antitumorais da combinação parecem ser mais influentes da reposição de p19Arf do que da expressão de IFN?, mas já in vivo, na presença do sistema imune, foram mais dependentes do IFN?. Com a combinação estes efeitos mostraram-se mais pronunciados, induzindo uma proteção antitumoral e maior sobrevida aos animais vacinados. Estes resultados indicam que a combinação p19Arf e IFN? pode ser aplicada como um agente imunoterápico e sugerem que a associação entre morte celular e imuno estimulação pode beneficiar o tratamento contra o câncer / Previously, we have shown in a mouse melanoma model of in situ gene therapy that co-transduction, but not individual application, with adenovirus vectors expressing the Interferon-beta (IFN?) (immune modulatory cytokine) and p19Arf (functional partner of the p53 tumor suppressor) transgenes results in massive cell death and reduced tumor progression. However, the capability of this combined treatment to stimulate an antitumor immune response has not been evaluated. Therefore, the aim of this work was to investigate, trough a prophylactic vaccine model, if B16F10 cells treated by the p19Arf and IFN? combination could induce such immune response. To do so, these cells were co-transduced by the AdPGp19 e AdPGIFN? adenoviral vectors and 48 hours after, inoculated as a vaccine agent in the left flank (vaccine site) of immune competent C57BL/6 mice. Seven days after the last vaccine, a tumor challenge was done with naïve B16F10 cells in the right flank (challenge site). Tumor progression was markedly reduced, even when challenge was done 73 days after the vaccination. However, since these animals developed tumors where the vaccine was applied, more appropriate conditions for the use of these treated cells were pursued, thus revealing that: the number of cells and inoculations can dictate tumor development, and also, that only with the combined treatment was tumor formation abolished. The influence of the immune system for this result was revelead by performing an immune supression protocol. Next, the roles of p19Arf and of IFN? were studied. In vitro, the antitumor effects were stronger upon the introduction of p19Arf than IFN?, but in vivo, in the presence of the immune system, the effects were more IFN? dependent. In fact, these effects were more pronouced with the combined treatment, inducing protection against tumor formation and progression and increasing survival in the vaccinated animals. Taken together, these results demonstrate the application of cells treated by the p19Arf e IFN? combination as an effective vaccine agent and also indicates that the association between cell death and immune stimulation may benefit the treatment of cancer
22

Células-tronco mensequimais como carreadoras de adenovírus no microambiente tumoral / Mesenchymal stem cell as carrier of adenovirus in the tumor microenvironment

Costa, Ruana Calado da 02 May 2017 (has links)
As muitas formas diferentes de câncer representam uma grande dimensão no âmbito da saúde pública mundial. Embora os esforços da medicina diagnóstica, vários tumores permanecem sem resposta à terapia tradicional. Uma alternativa é o uso de terapia gênica, a qual consiste a transferência de um gene terapêutico para a célula tumoral com a expectativa de inibição da progressão tumoral. Nosso laboratório desenvolveu uma série de vetores adenovirais onde a expressão do transgene é controlada pela p53 e usamos esses vetores para mostrar que a presença de p19Arf (um parceiro funcional de p53) sensibiliza células de melanoma murino, B16-F10 (p53-tipo selvagem), associado à ação do interferão-beta (IFNbeta, uma citocina pleiotrópica) quando testado in vitro. Mesmo que os vetores adenovirais representem o sistema de transferência gênica mais utilizado para a terapia de genes de câncer, seu uso por via sistêmica é limitado principalmente por inativação pelo sistema imune. Diferentes técnicas visam proteger as partículas de vírus do sistema imunológico e direcioná-las para o leito tumoral. Uma dessas técnicas envolve a utilização de células estaminais mesenquimais (MSCs). As propriedades dos MSC incluem a auto renovação, o potencial de diferenciação, bem como a sua capacidade de migrar e infiltrar tumores. Para este fim, nosso objetivo era utilizar MSCs murinos como portadores de adenovírus que expressam IFNbeta e para verificar se a presença de p19Arf nas células tumorais aumentaria a sua sensibilidade para IFNbeta. Para itso, os CTMs foram isolados da medula óssea ou do tecido adiposo de ratinhos C57BL/ 6 machos. Foi verificada a presença de marcadores de CTM (Sca1, CD29) e a ausência de marcadores para linhagens hematopoiéticas (CD31, CD11b, CD45). Sendo as CTM do tecido adiposo foram mais fáceis de cultivar, estes foram utilizados nos seguintes ensaios. In vitro, a aplicação do vector adenoviral que codifica um gene repórter (eGFP) resultou em mais de 70% de eficiênciamde transdução de CTM, sem indução de alterações morfológicas até 72 horas após o tratamento. A aplicação de vector portador de IFNbeta também foi bem tolerada, no entanto transdução com p19Arf sozinho ou em combinação com IFNbeta induziu alterações morfológicas nas CTMs. Em seguida, as células B16-F10 foram transduzidas ou não com o vetor codificando p19Arf e co-cultivadas com MSCs que foram transduzidas ou não com IFNbeta, demonstrando que a presença de p19Arf confere sensibilidade aumentada de células B16-F10 ao tratamento com IFNbeta . Em ensaios preliminares, os tumores B16-F10 foram estabelecidos subcutaneamente em camundongos C57BL / 6 e, posteriormente, as MSC marcadas com eGFP foram aplicadas na circulação após a injeção da veia da cauda. Após 48 horas, estes tumores foram recuperados e a presença de células positivas para eGFP foi confirmada, indicando que os MSCs se infiltraram no microambiente do tumor / The many different forms of cancer represent a tremendous investment for public health all over the world. Although the efforts of both diagnostic and therapeutic medicine have reduced the number of deaths due to cancer, many tumor types remain impervious to traditional therapy. An alternative is the use of gene therapy which entails the transfer of a therapeutic gene to the tumor cells with the expectation of inhibiting tumor progression. Our laboratory has developed a series of adenoviral vectors where transgene expression is controlled by p53 and we have used these vectors to show that the presence of p19Arf (a functional partner of p53) sensitizes murine melanoma cells, B16-F10 (p53-wild type), to the action of interferon-beta (IFNbeta, a pleiotropic cytokine) when tested in vitro. Even though adenoviral vectors are the most utilized gene transfer system for cancer gene therapy, their systemic application is limited principally by immune inactivation. Different techniques aim to protect the virus particles from the immune system and to direct them to the tumor bed. One of these techniques involves the utilization of mesenchymal stem cells (MSCs). The properties of MSCs include self-renewal, the potential for differentiation as well as their ability to migrate to and infiltrate tumors. To this end, our objective was to utilize murine MSCs as carriers of adenovirus that express IFNbeta and to verify if the presence of p19Arf in the tumor cells would enhance their sensitivity to IFNbeta. For this, MSCs were isolated from bone marrow or adipose tissue from male C57BL/6 mice. The presence of MSC markers (Sca1, CD29) was verified as was the absence of markers for hematopoietic lineages (CD31, CD11b, CD45). Since the MSCs from adipose tissue were easier to cultivate, these were utilized in the following assays. In vitro, application of the adenoviral vector encoding a reporter gene (eGFP) at a multiplicity of infection of 1000 resulted in the transduction of more than 70% of the MSCs and without the induction of morphological alterations even by 72 hours post treatment. The application of a vector encoding IFN? was also well tolerated, however transduction with p19Arf alone or in combination with IFNbeta induced morphologic alterations in the MSCs. Next, B16-F10 cells were transduced or not with the vector encoding p19Arf and co-cultivated with MSCs that had been transduced or not with IFNbeta, demonstrating that the presence of p19Arf confers enhanced sensitivity of B16-F10 cells to the treatment with IFN?. In preliminary assays, B16-F10 tumors were established subcutaneously in C57BL/6 mice and later MSCs labeled with eGFP were applied in the circulation upon tail vein injection. After 48 hours, these tumors were recovered and the presence of eGFP-positive cells was confirmed, indicating that the MSCs infiltrated the tumor microenvironment
23

Células-tronco mensequimais como carreadoras de adenovírus no microambiente tumoral / Mesenchymal stem cell as carrier of adenovirus in the tumor microenvironment

Ruana Calado da Costa 02 May 2017 (has links)
As muitas formas diferentes de câncer representam uma grande dimensão no âmbito da saúde pública mundial. Embora os esforços da medicina diagnóstica, vários tumores permanecem sem resposta à terapia tradicional. Uma alternativa é o uso de terapia gênica, a qual consiste a transferência de um gene terapêutico para a célula tumoral com a expectativa de inibição da progressão tumoral. Nosso laboratório desenvolveu uma série de vetores adenovirais onde a expressão do transgene é controlada pela p53 e usamos esses vetores para mostrar que a presença de p19Arf (um parceiro funcional de p53) sensibiliza células de melanoma murino, B16-F10 (p53-tipo selvagem), associado à ação do interferão-beta (IFNbeta, uma citocina pleiotrópica) quando testado in vitro. Mesmo que os vetores adenovirais representem o sistema de transferência gênica mais utilizado para a terapia de genes de câncer, seu uso por via sistêmica é limitado principalmente por inativação pelo sistema imune. Diferentes técnicas visam proteger as partículas de vírus do sistema imunológico e direcioná-las para o leito tumoral. Uma dessas técnicas envolve a utilização de células estaminais mesenquimais (MSCs). As propriedades dos MSC incluem a auto renovação, o potencial de diferenciação, bem como a sua capacidade de migrar e infiltrar tumores. Para este fim, nosso objetivo era utilizar MSCs murinos como portadores de adenovírus que expressam IFNbeta e para verificar se a presença de p19Arf nas células tumorais aumentaria a sua sensibilidade para IFNbeta. Para itso, os CTMs foram isolados da medula óssea ou do tecido adiposo de ratinhos C57BL/ 6 machos. Foi verificada a presença de marcadores de CTM (Sca1, CD29) e a ausência de marcadores para linhagens hematopoiéticas (CD31, CD11b, CD45). Sendo as CTM do tecido adiposo foram mais fáceis de cultivar, estes foram utilizados nos seguintes ensaios. In vitro, a aplicação do vector adenoviral que codifica um gene repórter (eGFP) resultou em mais de 70% de eficiênciamde transdução de CTM, sem indução de alterações morfológicas até 72 horas após o tratamento. A aplicação de vector portador de IFNbeta também foi bem tolerada, no entanto transdução com p19Arf sozinho ou em combinação com IFNbeta induziu alterações morfológicas nas CTMs. Em seguida, as células B16-F10 foram transduzidas ou não com o vetor codificando p19Arf e co-cultivadas com MSCs que foram transduzidas ou não com IFNbeta, demonstrando que a presença de p19Arf confere sensibilidade aumentada de células B16-F10 ao tratamento com IFNbeta . Em ensaios preliminares, os tumores B16-F10 foram estabelecidos subcutaneamente em camundongos C57BL / 6 e, posteriormente, as MSC marcadas com eGFP foram aplicadas na circulação após a injeção da veia da cauda. Após 48 horas, estes tumores foram recuperados e a presença de células positivas para eGFP foi confirmada, indicando que os MSCs se infiltraram no microambiente do tumor / The many different forms of cancer represent a tremendous investment for public health all over the world. Although the efforts of both diagnostic and therapeutic medicine have reduced the number of deaths due to cancer, many tumor types remain impervious to traditional therapy. An alternative is the use of gene therapy which entails the transfer of a therapeutic gene to the tumor cells with the expectation of inhibiting tumor progression. Our laboratory has developed a series of adenoviral vectors where transgene expression is controlled by p53 and we have used these vectors to show that the presence of p19Arf (a functional partner of p53) sensitizes murine melanoma cells, B16-F10 (p53-wild type), to the action of interferon-beta (IFNbeta, a pleiotropic cytokine) when tested in vitro. Even though adenoviral vectors are the most utilized gene transfer system for cancer gene therapy, their systemic application is limited principally by immune inactivation. Different techniques aim to protect the virus particles from the immune system and to direct them to the tumor bed. One of these techniques involves the utilization of mesenchymal stem cells (MSCs). The properties of MSCs include self-renewal, the potential for differentiation as well as their ability to migrate to and infiltrate tumors. To this end, our objective was to utilize murine MSCs as carriers of adenovirus that express IFNbeta and to verify if the presence of p19Arf in the tumor cells would enhance their sensitivity to IFNbeta. For this, MSCs were isolated from bone marrow or adipose tissue from male C57BL/6 mice. The presence of MSC markers (Sca1, CD29) was verified as was the absence of markers for hematopoietic lineages (CD31, CD11b, CD45). Since the MSCs from adipose tissue were easier to cultivate, these were utilized in the following assays. In vitro, application of the adenoviral vector encoding a reporter gene (eGFP) at a multiplicity of infection of 1000 resulted in the transduction of more than 70% of the MSCs and without the induction of morphological alterations even by 72 hours post treatment. The application of a vector encoding IFN? was also well tolerated, however transduction with p19Arf alone or in combination with IFNbeta induced morphologic alterations in the MSCs. Next, B16-F10 cells were transduced or not with the vector encoding p19Arf and co-cultivated with MSCs that had been transduced or not with IFNbeta, demonstrating that the presence of p19Arf confers enhanced sensitivity of B16-F10 cells to the treatment with IFN?. In preliminary assays, B16-F10 tumors were established subcutaneously in C57BL/6 mice and later MSCs labeled with eGFP were applied in the circulation upon tail vein injection. After 48 hours, these tumors were recovered and the presence of eGFP-positive cells was confirmed, indicating that the MSCs infiltrated the tumor microenvironment
24

Dissecting the Role of a lncRNA and Involvement of <em>Plasmodium</em> Infections in the Innate Immune Response: A Dissertation

Chan, Jennie 14 April 2015 (has links)
The innate immune system is a multicomponent response governed by intricate mechanisms of induction, regulation and resolution to elicit antimicrobial defenses. In recent years, the complexity of eukaryotic transcriptomes has become the subject of intense scrutiny and curiosity. It has been established, that RNA polymerase II (RNAPII) transcribes hundreds to thousands of long noncoding RNAs (lncRNAs), often in a stimulus and cell-type specific manner. However, the functional significance of these transcripts has been particularly controversial. While the number of identified lncRNAs is growing, our understanding of how lncRNAs themselves regulate other genes is quite limited. In chapter 2, a novel lncRNA is identified, more specifically, a natural antisense transcript, that mediates the transcription of the pro-inflammatory cytokine IL-1α. Through loss-of-function studies, I report the necessity of this transcript in mediating IL-1α mRNA expression by affecting RNAPII binding to the IL-1α promoter after toll-like receptor signaling. For the first time, I show that IL-1α is regulated at the transcriptional level. As a second independent component of this thesis, we explore the role of the innate immune response after infection by the malaria-causing parasite, Plasmodium berghei ANKA (PbA), and how innate immune components are both beneficial and detrimental to the host depending on when and where inflammation is triggered during infection. We attempt to identify the “malarial toxin” responsible for aberrations in the immune response that is detrimental for disease outcomes and the innate signaling pathways that are involved. Many pathogens induce pathological inflammatory conditions that lead to irreparable homeostatic imbalances and become fatal to the host. Here, type I Interferon signaling is required to dampen parasite load during liver-stage infections, but leads to host mobidity if these pathways are activated in the erythrocytic phase of infection. Together, this thesis provides new insights on how components of the innate immune system are regulated, and how dysregulation of immunity can potentially lead to adverse effects during active infections.

Page generated in 0.0949 seconds