• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 24
  • 19
  • 13
  • 2
  • 2
  • 1
  • 1
  • Tagged with
  • 68
  • 41
  • 17
  • 15
  • 14
  • 14
  • 13
  • 12
  • 12
  • 12
  • 12
  • 11
  • 10
  • 9
  • 9
  • 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.
31

Identificação da família BCL2 como alvo terapêutico no tratamento das neoplasias mieloproliferativas associadas à mutação da JAK2V617F / BCL2 family as potential therapeutical targets in the treatment of JAK2V617F- associated myeloproliferative neoplasms

Cristina Tavares Leal 01 September 2017 (has links)
As neoplasias mieloproliferativas (NMPs) negativas para o rearranjo t(9;22)/BCRABL1, incluindo Policitemia Vera (PV), Trombocitemia Essencial (TE) e Mielofibrose Primária (MFP), são doenças hematopoéticas clonais e estão frequentemente associadas à mutação JAK2V617F. Apesar dos avanços no conhecimento da fisiopatologia após a descoberta da mutação JAK2V617F e do desenvolvimento de inibidores da JAK2, o tratamento permanece não curativo. Sabe-se que as célulastronco mais primitivas nas NMPs são responsáveis pela iniciação da doença e que a expansão dos precursores mieloeritróides contribui para o fenótipo clínico. Dados recentes obtidos com ensaios in vitro mostram que as proteínas da família BCL2, reguladoras da apoptose mitocondrial, desempenham um papel relevante na patogênese das NMPs. Acreditamos que a expressão anômala de BCL2 nas células progenitoras hematopoéticas (CPH) das NMPs pode contribuir para a patogênese desse grupo de doenças. Avaliamos a expressão gênica, por meio de PCR em Tempo Real, da família BCL2 (genes antiapoptóticos BCL-xL e BCL2 e o pró-apoptótico BIM) nas diferentes subpopulações de progenitores hematopoéticos murinos (de um modelo condicional knockin de expressão heterozigótica condicional da Jak2V617F) e de pacientes portadores de NMPs bem como sua contribuição para o fenótipo da doença e resposta ao inibidores da JAK2 (com a droga ruxolitinibe) e/ou inibição da família BCL2 (com o inibidor de BCL2 obatoclax). Não encontramos diferença de expressão basal dos genes BCL2, BCL-xL e BIM nas células CD34+ bem como nas subpopulações de células CD34+38-/+ de pacientes com NMPs, independente da presença da mutação JAK2V617F, em relação às células CD34+ e subpopulações CD34+38-/+ dos controles (p>0.05). Nas células CD34+ de pacientes com TE encontramos aumento de expressão de BCL2 em relação às células CD34+ pacientes com MFP (p=0.03). No modelo transgênico de camundongos Jak2 wt/VF (que apresentam uma NMP semelhante à PV) e Jak2 wt/wt (controles), comparamos a expressão diferencial dos genes da família Bcl2 em precursores hematopoéticos imaturos (LSKs) e progenitores mieloides mais maduros (MPs). A expressão do BclxL em MPs de camundongos wt/VF foi maior em relação à subpopulação de células LSKs e em relação as duas subpopulações de células dos controles (p=0.0011). Não houve diferença significativa de expressão do Bcl2 nas subpopulações de células LSKs e MPs de animais wt/VF e wt/wt (p=0.12). Observou-se menor expressão de Bim em LSKs em relação às células MPs dos animais mutados (p=0.026), diferença essa não observada entre os controles Jak2 wt/wt. O tratamento isolado com inibidor de JAK2 ou de BCL2 resultou em aumento de expressão do Bim nas CPH (LSKs e MPs) de camungongos Jak2 wt/VF em relação aos animais Jak2 wt/wt. Este aumento da expressão de Bim foi ainda mais evidente após o tratamento das células com a combinação das duas drogas quando comparadas às células não tratadas ou tratadas com um dos dois inibidores, sendo maior em animais doentes do que em animais controles (p<0.0001). A análise do efeito do tratamento com os inibidores de JAK2 e BCL2 na indução de apoptose por meio de citometria de fluxo (marcação com anexina/7-AAD) revelou que as células LSKs foram mais resistentes à apoptose tardia do que as células MPs independentemente da mutação da JAK2 (p<0.05). O tratamento com obatoclax resultou em indução de apoptose diferentemente do que foi observado com o tratamento com ruxolitinibe (p=0.594) nas células MPs de animais Jak2 wt/VF. Ademais, o tratamento combinado com ruxolitinibe e obatoclax resultou no aumento da apoptose nas células MPs dos animais com fenótipo de PV (Jak2 wt/VF) em relação aos animais Jak2 wt/wt (p=0.05). Em conclusão, demonstramos que a resistência à apoptose nas NMPs ocorre desde as CPH iniciadoras da doença. Nossos resultados sugerem que a modulação da apoptose mitocondrial pode ser uma nova estratégia terapêutica para pacientes com NMP em combinação aos inibidores de JAK2, na medida em que atua tanto nas CPH que iniciam a doença como nos MPs, responsáveis pelos sinais e sintomas de mieloproliferação. / Myeloproliferative Neoplasms (MPNs) negative for t(9;22)/BCR-ABL1 rearrangement, including Polycythemia Vera (PV), Essential Thrombocythemia (ET) and Primary Myelofibrosis (PMF), are clonal hematopoietic diseases and are often associated with the JAK2V617F mutation. Despite advances in the pathophysiology knowledge after the discovery of the JAK2V617F mutation and the development of JAK2 inhibitors, treatment remains non-curative. It is known that MPN primitive stem cells are essential for the initiation of the disease and that the expansion of the myeloeritroid precursors contributes to the clinical phenotype. Recent data, obtained with in vitro assays, showed that BCL2 family proteins, regulators of mitochondrial apoptosis, play a relevant role in the pathogenesis of MPNs. We believe that the anomalous expression of BCL2 in hematopoietic progenitor cells (HPCs) of MPNs may contribute to their pathogenesis. We evaluated BCL2 family (antiapoptotic genes BCL-xL and BCL2 and the pro-apoptotic BIM) gene expression by real-time PCR in different subpopulations of hematopoietic progenitors from a conditional Jak2V617F knockin murine model and from patients with MPNs as well as their contribution to the disease phenotype and response to JAK2 inhibitors (with ruxolitinib) and/or to the inhibition of the BCL2 family (with the BH3-mimetic obatoclax). We found no difference in the basal expression of the BCL2, BCL-xL and BIM in CD34+ cells as well as in subpopulations of CD34+ 38-/+ cells from patients with MPNs, regardless of the presence of the JAK2V617F mutation. In CD34+ cells obtained from patients with ET, we found an increase of BCL2 expression when compared to CD34+ cells with PMF (p=0.03). In the Jak2 wt/VF transgenic mice (that develop a MPN similar to PV) and Jak2 wt/wt controls, we compared the differential expression of Bcl2 family genes in immature hematopoietic precursors (LSKs) and more mature myeloid progenitors (MPs). Expression of Bcl-xL in MPs of wt/VF mice was greater when compared to LSKs and to the two progenitor subpopulations of control cells (p=0.0011). There was no significant difference in Bcl2 expression between the subpopulations of LSKs and MPs from wt/VF and wt/wt animals (p=0.12). Lower Bim expression in LSKs than in MPs was observed in samples from JAK2-mutated animals (p=0.026). Such difference was not observed between the Jak2 wt/wt subpopulations. Treatment with JAK2 or BCL2 inhibitors alone resulted in increased Bim expression in LSKs and MPs of the Jak2 wt/VF mice when compared to Jak2 wt/wt animals. This increase in Bim expression was even more evident when these cells were treated with the combination of the two drugs as compared to single treatment with one of the two inhibitors, being higher in mutaded than control animals (p<0.0001). The analysis of apoptosis by flow cytometry (annexin / 7-AAD labeling) revealed that LSK cells were more resistant to late apoptosis than MP cells regardless of the JAK2 mutation (p<0.05). Treatment with obatoclax resulted in greater apoptosis induction than it was observed with ruxolitinib treatment (p=0.594) on MP cells of Jak2 wt/VF animals. In addition, the combined treatment with ruxolitinib and obatoclax resulted in increased apoptosis in MP cells of animals with the PV phenotype (Jak2 wt/VF) as compared to the Jak2 wt/wt animals (p=0.05). In conclusion, we demonstrated that resistance to apoptosis in MPNs occurs at the level of the hematopoietic progenitors that initiate the disease. Our results suggest that modulation of mitochondrial apoptosis may be a new therapeutic strategy for MPN patients in combination with JAK2 inhibitors, as it acts on both the disease initiating and more mature progenitors, responsible for the clinical findings of myeloproliferation.
32

Expressão de Galectinas-1 e 3 em Neoplasias Mieloproliferativas / Galectin-1 and 3 expression in Myeloproliferative Neoplasms

Lívia Gonzaga Moura 26 October 2012 (has links)
Doenças Mieloproliferativas Crônicas são desordens hematológicas malignas caracterizadas pela alteração na célula-tronco hematopoética e independência ou hipersensibilidade dos progenitores hematopoéticos a citocinas. Em 2008 a OMS renomeou esse grupo como Neoplasias Mieloproliferativas (NMPs), no qual estão inclusas as entidades nosológicas Policitemia Vera (PV), Trombocitemia Essencial (TE) e Mielofibrose Primária (MFP), doenças alvo desse estudo. Apesar dos avanços no diagnóstico das NMPs e nos mecanismos envolvidos com a fisiopatologia dessas doenças, sua patogênese permanece desconhecida. Alterações na maquinaria apoptótica parecem estar envolvidas em na fisiopatologia das NMP e por isso a compreensão dos mecanismos de regulação da apoptose e a interferência das galectinas-1 e 3 nesse processo, em pacientes com NMPs, é relevante para a busca de novos alvos terapêuticos. Neste contexto, os objetivos deste trabalho foram: avaliar em leucócitos de sangue periférico e células tronco hematopoéticas CD34+ de medula óssea dos pacientes com PV, TE e MFP os níveis de expressão das LGALS1 e LGALS3 e a concentração de galectina-3 plasmática. Foram determinadas as correlações dos níveis de expressão de LGALS1 e LGALS3 e da concentração da galectina-3 plasmática com os níveis de expressão do RNAm das moléculas reguladoras da apoptose e com os dados clínico-laboratoriais dos pacientes como a concentração de hemoglobina, percentagem de hematócrito, porcentagem de alelos mutados JAK2V617F, contagem de leucócitos e esplenomegalia. A expressão de LGALS1 estava diminuída em células CD34+ em PV e MFP e em leucócitos de sangue periférico de pacientes com MFP. Os pacientes de TE apresentaram aumento na expressão de LGALS3 em leucócitos de sangue periférico e alta concentração de galectina-3 no plasma. Houve correlação entre os níveis de expressão de LGALS1 e a porcentagem de alelos mutados e a contagem de leucócitos, em pacientes com PV. Foi detectada a correlação entre os níveis de expressão de LGALS3 a porcentagem de alelos mutados e o tamanho do baço, em pacientes com MFP. Com relação aos genes reguladores da apoptose, foram observadas correlações entre os níveis de expressão de LGALS1 e BCL-2 em células CD34+ de pacientes PV e entre LGALS3 e A1, MCL-1, BAX e C-FLIP em leucócitos de de pacientes com TE. Os resultados obtidos indicam que as NPM apresentam expressão diferencial de LGALS1 e LGALS3 e sugerem a associação entre a expressão de galectinas e o status da mutação JAK2V617F, principalmente em pacientes com MFP / Chronic myeloproliferative diseases are haematological malignant disorders characterized by the presence of an altered haematopoietic stem cell and independence or hypersensibility of their hematopoietic progenitors to cytokines. In 2008, WHO renamed this group of diseases as Myeloproliferative Neoplasms (MPN) in which is included Polycythemia Vera (PV), Essential Thrombocythemia (ET) and Primary Myelofibrosis (PMF). There have been advances concerning the knowledge about the mechanisms involved in MPN pathophysiology, however their pathogenesis remains unknow. Deregulation in apoptotic machinery seems to be involved in MPN pathophysiology. Fully understanding of apoptotic machinery and the influence of galectin-1 and 3 in this process in NMP patients might unveil novel targets for manipulation. The aims of the present study were to evaluate in leukocytes and CD34+ hematopoietic stem cells from PV, ET and PMF patients the LGALS1 and LGALS3 expression levels, the Galectin-3 plasma levels and to correlate LGALS1 and LGALS3 expression levels with galectin-3 plasma levels, apoptosis-related genes expression, JAK2 mutation status and clinic-laboratorial parameters. PV and PMF patients showed decreased expression levels of LGALS1 in CD34+ cells and also decreased LGALS1 expression levels in PMF leukocytes. ET patients presented an increased expression level of galectin-3 in leukocytes and plasma. We detected the correlations between LGALS3 gene expression with JAK2 allele burden and with leukocytes number in PV patients. We also observed in PMF patients the correlation between LGALS3 expression levels with JAK2 allele burden and spleen size. We also detected the correlation between LGALS1 expression levels BCL-2 gene expression in PV CD34+ HSC cells and between LGALS3 expression and A1, MCL-1, BAX and C-FLIP gene expression in TE leukocytes. Taken together, the results suggest the LGALS1and LGALS3 differential expression in NMP and the relation between JAK2V617F status with galectins expression, especially in PMF patients.
33

Caracterização do papel das células Natural Killer nas neoplasias mieloproliferativas / Characterization of the Natural Killer cells role in myeloproliferative neoplasms

Adriana Queiroz Arantes Rocha 19 October 2017 (has links)
As células Natural Killer (NK), quando estimuladas por meio de seus receptores, rapidamente produzem citocinas e quimiocinas, incluindo IFN?, TNF?, TGF?, GMCSF, MIP1?, MIP1?, IL-10 e outras, as quais podem afetar a função de outras células hematopoéticas. Considerando as evidências recentes de que as célulastronco hematopoéticas (CTH) respondem diretamente à sinalização de várias citocinas, acreditamos que a produção de citocinas mediada pelas células NK possa regular a função da CTH e que a sua desregulação possa favorecer a transformação maligna. As neoplasias mieloproliferativas (NMP) negativas para o rearranjo t(9;22)/BCR-ABL1, incluindo as entidades Policitemia Vera (PV), Trombocitemia Essencial (TE) e Mielofibrose Primária (MFP), são doenças hematopoéticas originadas de alteração clonal da CTH, e podem servir de modelo para o estudo dessa regulação NK-CTH. Além de mutações que ativam vias de proliferação e sobrevivência celular, como a mutação JAK2V617F, presente em mais da metade das NMP, outros mecanismos contribuem para a patogênese e manutenção da doença, tais como mutações adicionais e regulação da hematopoese neoplásica pelo microambiente da medula óssea. Este último inclui não apenas células do estroma, mas também células do sistema imune. Dessa forma, com objetivo de investigar a potencial contribuição das células NK para a patogênese das NMP, caracterizamos células do sangue periférico de pacientes com NMP do Ambulatório de Hematologia do Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto, bem como células esplênicas obtidas de animais de um modelo murino condicional knockin de expressão heterozigótica da Jak2V617F (Jak2VF) quanto à frequência, expressão de receptores e função das células NK. Observamos menor porcentagem de células NK e maior expressão do receptor inibitório NKG2A nos animais Jak2 wt/VF. Pacientes portadores de NMP apresentaram número absoluto de células NK-CD16+ reduzido em relação aos controles saudáveis. O número de células NK-CD16+ foi menor na MFP em relação aos controles e à TE, particularmente naqueles portadores da mutação JAK2V617F. Encontramos menor expressão do receptor de ativação NKG2D nos portadores de PV positivos para a mutação JAK2V617F. Houve menor expressão do receptor de ativação NKp46 nos portadores de NMP, particularmente nos portadores da mutação JAK2V617F, e nos pacientes com MFP. Observamos também menor expressão do receptor NKG2A nos portadores de TE negativos para a mutação JAK2V617F. Em concordância com a redução de células NK, a porcentagem de linfócitos totais mostrou-se reduzida nos pacientes com NMP, particularmente nos portadores de MFP, independentemente da mutação JAK2V617F. Encontramos redução percentual e absoluta do subtipo de células NK CD56brightCD16- (cuja principal função é secretória) nos pacientes com NMP, especialmente na presença da mutação JAK2V617F, nos pacientes com PV e MFP, e menor frequência absoluta do subtipo CD56-CD16bright (com função primariamente citotóxica) nos portadores de MFP. Em contraste, não verificamos deficiência citotóxica das células NK dos animais Jak2 wt/VF em relação aos controles Jak2 wt/wt. Adicionalmente, as células NK dos animais Jak2-mutados demonstraram menor capacidade de secreção da citocina MIP-1?, reconhecida por regular a função de CTH, em relação aos animais controle. Finalmente, houve expressão significativamente aumentada do gene MyD88 nos animais mutados em relação aos controles, sugerindo que a via de sinalização dos receptores do tipo Toll (TLR) pode estar envolvida na regulação NKCTH nas NMP. Em resumo, detectamos deficiência numérica e funcional de células NK em células primárias murinas e humanas de NMP. Nossos achados sugerem potencial regulação da hematopoese maligna pelas células NK nestas neoplasias e podem contribuir para a identificação de novas estratégias terapêuticas que possam interferir nesta complexa interação. / Natural Killer (NK) cells, when stimulated by their receptors, rapidly produce cytokines and chemokines, including IFN?, TNF?, TGF?, GM-CSF, MIP1?, MIP1?, IL-10 and others, which may affect the function of other hematopoietic cells. Considering the recent evidence that hematopoietic stem cells (HSC) directly respond to cytokine signaling, we hypothesized that NK cells mediated cytokine production can regulate HSC function and that their dysregulation may favor malignant transformation. BCR-ABL1-negative myeloproliferative neoplasms (MPN), including Polycythemia Vera (PV), Essential Thrombocythemia (ET) and Primary Myelofibrosis (PMF), are hematopoietic diseases originated from HSC clonal transformation, and thus can serve as a model for studying this NK-HSC regulation. In addition to mutations that activate cell proliferation and survival pathways, such as the JAK2V617F mutation, present in more than half of MPN cases, other mechanisms contribute to the pathogenesis and maintenance of the disease, such as additional mutations and regulation of neoplastic hematopoiesis by the bone marrow microenvironment. This latter includes not only stromal cells but also cells of the immune system. Therefore, in order to investigate the potential contribution of NK cells to the pathogenesis of MPN, we characterized the frequency, receptor expression and function of NK cells from patients with MPN from the Clinical Hospital of the Medical School of Ribeirão Preto, University of São Paulo, as well as from splenic cells obtained from animals of a conditional knockin murine model of Jak2V617F heterozigous expression. Lower percentage of NK cells and higher NKG2A inhibitory receptor expression was observed in Jak2 wt/VF animals as compared to Jak2 wt/wt controls. In agreement, patients with MPN presented reduced absolute numbers of NK-CD16+ cells when compared to healthy controls. The number of NKCD16+ cells was lower in the PMF than in controls or ET patients, particularly in those bearing the JAK2V617F mutation. We found lower expression of the NKG2D activatory receptor in PV patients with the JAK2V617F mutation. There was lower expression of the NKp46 activatory receptor in MPN patients, particularly in those with the JAK2V617F mutation, and in PMF patients. We also observed reduced expression of the NKG2A receptor in non-JAK2 mutated ET patients. In agreement with the NK cell reduction, the percentage of total lymphocytes was reduced in patients with MPN, particularly in PMF, regardless of the JAK2V617F mutation. We found an absolute decrease of the CD56brightCD16- NK subtype (whose main function is secretory) in patients with MPN, especially when the JAK2V617F mutation was present, in patients with PV and PMF. Also, lower absolute frequency of CD56-CD16bright NK subtype (primarily cytotoxic) was found in PMF patients. In contrast, we did not find cytotoxic deficiency in the Jak2 wt/VF NK cells as compared to the Jak2 wt/wt controls. In addition, Jak2-mutated NK cells presented reduced ability of secreting the cytokine MIP-1?, known to regulate HSC function. Finally, there was significantly increased expression of the MyD88 gene in the Jak2-mutated animals as compared to controls, suggesting that the Toll-like receptors (TLR) signaling pathway may be involved in NK-HSC regulation in MPN. In summary, we detected numerical and functional deficiency of NK cells in murine and human primary cells of MPN. Our findings suggest a potential regulation of malignant hematopoiesis by NK cells in these neoplasms and may contribute to the identification of new therapeutic strategies that may target this complex interaction.
34

Inibição farmacológica dos substratos do receptor de insulina em neoplasia mieloproliferativa JAK2V617F / Pharmacological inhibition of insulin receptor substrates in myeloproliferative neoplasm JAK2V617F

Bruna Alves Fenerich 29 June 2017 (has links)
A mutação recorrente JAK2V617F é a lesão molecular com maior impacto na fisiopatologia das neoplasias mieloproliferativas (NMP) BCR ABL1 negativas. A ausência de resposta clínica completa ao inibidor seletivo de JAK1/2, ruxolitinibe, indica a necessidade de novas abordagens terapêuticas. Dados recentes sugerem que IGF1R/IRS representa um potencial alvo de inibição para o tratamento das NMP: (i) o substrato do receptor de insulina 2 (IRS2) coopera com JAK2V617F na transformação maligna em NMP; (ii) a desregulação da via de sinalização de IGF1R induz NMP. O composto NT157 foi desenvolvido para inibir IRS1/2 e apresentou efeitos antineoplásicos em neoplasias sólidas. Os objetivos deste trabalho foram avaliar os efeitos celulares e moleculares do tratamento com o inibidor de IRS1/2, NT157, isolado e em combinação com ruxolitinibe, em NMP JAK2V617F. Células HEL e SET2 JAK2V617F foram tratadas com veículo, NT157 e/ou ruxolitinibe e submetidas à avaliação da viabilidade celular, apoptose, proliferação, clonogenicidade, ciclo celular, expressão gênica e/ou expressão/ativação proteica. Células primárias de pacientes com policitemia vera foram submetidos a tratamento com NT157 e avaliação de formação espontânea de colônias eritroides. O efeito do NT157 in vivo foi avaliado utilizando modelo de xenotransplante de células HEL em camundongos NSG. A análise estatística foi realizada através do teste ANOVA ou t de Student. Em células HEL e/ou SET2 JAK2V617F, o tratamento com NT157 promoveu redução da viabilidade, clonogenicidade e proliferação celular, aumentou a apoptose e resultou em parada do ciclo celular em G2/M (p?0,05). Exposição ao NT157 resultou em inibição da fosforilação de STAT3, STAT5 e ERK e na modulação da expressão de 23 oncogenes (CCND1, MYB e WT1) e genes supressores tumorais (CDKN1A, JUN e FOS) em células HEL (p?0,05). O tratamento combinado com ruxolitinibe não apresentou efeito potencializador, sendo que a redução da viabilidade nas condições de combinação corresponde ao efeito das monoterapias nas linhagens celulares avaliadas. Em células primárias de pacientes com policitemia vera (n=3), NT157 reduziu a formação espontânea de colônias eritroides (p?0,05). O tratamento in vivo com veículo ou NT157 na dose de 70mg/kg, 3 vezes por semana, via intraperitoneal, em modelos de xenotransplante com células HEL em camundongos NSG (n=5 para cada grupo) não apresentou efeitos antineoplásicos. Em conclusão, a inibição farmacológica de IRS1/2 apresentou efeitos antineoplásicos significativos em modelos de linhagens celulares e amostras primárias de pacientes com NMP JAK2V617F. A inibição farmacológica combinada de IRS1/2 e JAK1/2 não potencializou o efeito antineoplásico das monoterapias nos processos celulares investigados. Os resultados dos estudos in vivo em modelos de xenotransplante indicam a necessidade de estudos de farmacocinética e farmacodinâmica para o NT157. Os efeitos moleculares identificados permitiram uma melhor compreensão sobre os mecanismos de ação da droga NT157 em NMP. / The recurrent V617F mutation in JAK2 is a major contributor to the pathogenesis of BCR-ABL1 negative myeloproliferative neoplasms (MPN). Absence of complete clinical response to ruxolitinib, a JAK1/2 inhibitor, highlights the need for targeting other signaling pathways that contribute to JAK2. Recent data indicate that IGF1R/IRS is a potential target in MPN: (i) insulin receptor substrate 2 (IRS2) cooperates to malignant transformation induced by JAK2V617F, (ii) IGF1R signaling upregulation induces MNP phenotype. NT157 is a synthetic compound designed as IRS1/2 inhibitor and was able to induce anti-neoplastic effects in solid tumors. We, herein, aimed to characterize the molecular and cellular effects of NT157 treatment, combined or not with ruxolitinib, in MPN JAK2V617F. HEL and SET2 JAK2V617F cells were treated or not with vehicle, NT157 and/or ruxolitinib and submitted to evaluation of cell viability assay, apoptosis, proliferation, clonogenicity, cell cycle, gene expression and protein expression/activation. Primary cells from polycythemia vera (PV) patients (n=3) were exposed to NT157 treatment and evaluated for erythropoietin-independent colony formation. NT157 effects in vivo were evaluated in a xenograft model of leukemogenesis induced by HEL cells in NSG mice. Statistical analysis was performed using ANOVA or Student\'s t test. In MPN cell lines, NT157 treatment significantly decreased cell viability, clonogenicity and cell proliferation, increased apoptosis and cell cycle arrest in G2/M (all p<0.05). NT157 exposure resulted in inhibition of STAT3, STAT5 and ERK phosphorylation. NT157 also modulated the expression of 23 oncogenes (CCND1, MYB and WT1) and suppressor tumor genes (CDKN1A, FOS and JUN) in HEL cells (p?0.05). In both cell lines, the combined treatment, NT157 plus ruxolitinib, did not potentiate the effects of monotherapies. In primary cells from polycythemia vera patients, NT157 exposition reduced spontaneous erythroid colony formation (all p<0.05). In vivo treatment with vehicle or NT157 (70mg/kg intraperitoneal), three times a week, showed no antineoplastic effects in NSG mice transplanted with HEL cells (n = 5 for each group). In summary, the IRS1/2 pharmacological inhibitor NT157 displayed remarkable antineoplastic effects in JAK2V617F cells lines and MPN primary cells. The combined treatment of NT157 plus ruxolitinib did not present potentializing effects when compared to the monotherapy. The results of in vivo treatment using a xenograft model highlight the need for pharmacokinetic and pharmacodynamic studies for the NT157 compound. The molecular effects identified allowed a better understanding about the mechanisms of NT157 action in MPNs.
35

Caracterização do papel das células Natural Killer nas neoplasias mieloproliferativas / Characterization of the Natural Killer cells role in myeloproliferative neoplasms

Rocha, Adriana Queiroz Arantes 19 October 2017 (has links)
As células Natural Killer (NK), quando estimuladas por meio de seus receptores, rapidamente produzem citocinas e quimiocinas, incluindo IFN?, TNF?, TGF?, GMCSF, MIP1?, MIP1?, IL-10 e outras, as quais podem afetar a função de outras células hematopoéticas. Considerando as evidências recentes de que as célulastronco hematopoéticas (CTH) respondem diretamente à sinalização de várias citocinas, acreditamos que a produção de citocinas mediada pelas células NK possa regular a função da CTH e que a sua desregulação possa favorecer a transformação maligna. As neoplasias mieloproliferativas (NMP) negativas para o rearranjo t(9;22)/BCR-ABL1, incluindo as entidades Policitemia Vera (PV), Trombocitemia Essencial (TE) e Mielofibrose Primária (MFP), são doenças hematopoéticas originadas de alteração clonal da CTH, e podem servir de modelo para o estudo dessa regulação NK-CTH. Além de mutações que ativam vias de proliferação e sobrevivência celular, como a mutação JAK2V617F, presente em mais da metade das NMP, outros mecanismos contribuem para a patogênese e manutenção da doença, tais como mutações adicionais e regulação da hematopoese neoplásica pelo microambiente da medula óssea. Este último inclui não apenas células do estroma, mas também células do sistema imune. Dessa forma, com objetivo de investigar a potencial contribuição das células NK para a patogênese das NMP, caracterizamos células do sangue periférico de pacientes com NMP do Ambulatório de Hematologia do Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto, bem como células esplênicas obtidas de animais de um modelo murino condicional knockin de expressão heterozigótica da Jak2V617F (Jak2VF) quanto à frequência, expressão de receptores e função das células NK. Observamos menor porcentagem de células NK e maior expressão do receptor inibitório NKG2A nos animais Jak2 wt/VF. Pacientes portadores de NMP apresentaram número absoluto de células NK-CD16+ reduzido em relação aos controles saudáveis. O número de células NK-CD16+ foi menor na MFP em relação aos controles e à TE, particularmente naqueles portadores da mutação JAK2V617F. Encontramos menor expressão do receptor de ativação NKG2D nos portadores de PV positivos para a mutação JAK2V617F. Houve menor expressão do receptor de ativação NKp46 nos portadores de NMP, particularmente nos portadores da mutação JAK2V617F, e nos pacientes com MFP. Observamos também menor expressão do receptor NKG2A nos portadores de TE negativos para a mutação JAK2V617F. Em concordância com a redução de células NK, a porcentagem de linfócitos totais mostrou-se reduzida nos pacientes com NMP, particularmente nos portadores de MFP, independentemente da mutação JAK2V617F. Encontramos redução percentual e absoluta do subtipo de células NK CD56brightCD16- (cuja principal função é secretória) nos pacientes com NMP, especialmente na presença da mutação JAK2V617F, nos pacientes com PV e MFP, e menor frequência absoluta do subtipo CD56-CD16bright (com função primariamente citotóxica) nos portadores de MFP. Em contraste, não verificamos deficiência citotóxica das células NK dos animais Jak2 wt/VF em relação aos controles Jak2 wt/wt. Adicionalmente, as células NK dos animais Jak2-mutados demonstraram menor capacidade de secreção da citocina MIP-1?, reconhecida por regular a função de CTH, em relação aos animais controle. Finalmente, houve expressão significativamente aumentada do gene MyD88 nos animais mutados em relação aos controles, sugerindo que a via de sinalização dos receptores do tipo Toll (TLR) pode estar envolvida na regulação NKCTH nas NMP. Em resumo, detectamos deficiência numérica e funcional de células NK em células primárias murinas e humanas de NMP. Nossos achados sugerem potencial regulação da hematopoese maligna pelas células NK nestas neoplasias e podem contribuir para a identificação de novas estratégias terapêuticas que possam interferir nesta complexa interação. / Natural Killer (NK) cells, when stimulated by their receptors, rapidly produce cytokines and chemokines, including IFN?, TNF?, TGF?, GM-CSF, MIP1?, MIP1?, IL-10 and others, which may affect the function of other hematopoietic cells. Considering the recent evidence that hematopoietic stem cells (HSC) directly respond to cytokine signaling, we hypothesized that NK cells mediated cytokine production can regulate HSC function and that their dysregulation may favor malignant transformation. BCR-ABL1-negative myeloproliferative neoplasms (MPN), including Polycythemia Vera (PV), Essential Thrombocythemia (ET) and Primary Myelofibrosis (PMF), are hematopoietic diseases originated from HSC clonal transformation, and thus can serve as a model for studying this NK-HSC regulation. In addition to mutations that activate cell proliferation and survival pathways, such as the JAK2V617F mutation, present in more than half of MPN cases, other mechanisms contribute to the pathogenesis and maintenance of the disease, such as additional mutations and regulation of neoplastic hematopoiesis by the bone marrow microenvironment. This latter includes not only stromal cells but also cells of the immune system. Therefore, in order to investigate the potential contribution of NK cells to the pathogenesis of MPN, we characterized the frequency, receptor expression and function of NK cells from patients with MPN from the Clinical Hospital of the Medical School of Ribeirão Preto, University of São Paulo, as well as from splenic cells obtained from animals of a conditional knockin murine model of Jak2V617F heterozigous expression. Lower percentage of NK cells and higher NKG2A inhibitory receptor expression was observed in Jak2 wt/VF animals as compared to Jak2 wt/wt controls. In agreement, patients with MPN presented reduced absolute numbers of NK-CD16+ cells when compared to healthy controls. The number of NKCD16+ cells was lower in the PMF than in controls or ET patients, particularly in those bearing the JAK2V617F mutation. We found lower expression of the NKG2D activatory receptor in PV patients with the JAK2V617F mutation. There was lower expression of the NKp46 activatory receptor in MPN patients, particularly in those with the JAK2V617F mutation, and in PMF patients. We also observed reduced expression of the NKG2A receptor in non-JAK2 mutated ET patients. In agreement with the NK cell reduction, the percentage of total lymphocytes was reduced in patients with MPN, particularly in PMF, regardless of the JAK2V617F mutation. We found an absolute decrease of the CD56brightCD16- NK subtype (whose main function is secretory) in patients with MPN, especially when the JAK2V617F mutation was present, in patients with PV and PMF. Also, lower absolute frequency of CD56-CD16bright NK subtype (primarily cytotoxic) was found in PMF patients. In contrast, we did not find cytotoxic deficiency in the Jak2 wt/VF NK cells as compared to the Jak2 wt/wt controls. In addition, Jak2-mutated NK cells presented reduced ability of secreting the cytokine MIP-1?, known to regulate HSC function. Finally, there was significantly increased expression of the MyD88 gene in the Jak2-mutated animals as compared to controls, suggesting that the Toll-like receptors (TLR) signaling pathway may be involved in NK-HSC regulation in MPN. In summary, we detected numerical and functional deficiency of NK cells in murine and human primary cells of MPN. Our findings suggest a potential regulation of malignant hematopoiesis by NK cells in these neoplasms and may contribute to the identification of new therapeutic strategies that may target this complex interaction.
36

Identification et fonction de nouvelles mutations des récepteurs à la thrombopoïétine et à l’érythropoïétine dans les néoplasmes myéloprolifératifs et les érythrocytoses. / Identification and role of thrombopoietin and erythropoietin receptors mutations in myeloproliferative neoplasms and erythrocytosis

Pasquier, Florence 05 November 2015 (has links)
Le récepteur à l’érythropoïétine (EPOR) peut être muté dans les érythrocytoses congénitales tandis que des mutations de MPL, récepteur à la thrombopoiétine, sont observées dans certains néoplasmes myéloprolifératifs (NMP). L’érythrocytose congénitale touche exclusivement les progéniteurs érythroïdes et se traduit par une polyglobulie isolée. Des mutations d’EPOR sont décrites dans environ 12% des cas. La première partie de cette thèse reposait sur l’étude fonctionnelle d’une mutation d’EPOR, jamais décrite, c.1300dupC (p.Gln434Profs*11). Ce mutant est responsable, dans les cellules primaires et les lignées cellulaires, d’une hypersensibilité majeure à l’EPO qui n’est pas due à la perte de sites de régulation négative du signal ou à un défaut d’internalisation du récepteur, contrairement aux données de la littérature, mais à une stabilisation d’EPOR à la membrane cellulaire par probable changement conformationnel. Dans la seconde partie, un variant d’EPOR Pro488Ser a été étudié au sein d’une famille de NMP. Seule une activation spontanée faible de STAT5 a pu être mise en évidence dans les lignées cellulaires. Des modèles murins et/ou d’iPSC seront développés dans l’hypothèse d’une coopération entre EPOR P488S et JAK2V617F. Enfin, dans la dernière partie de ce travail, nous avons réalisé une étude fonctionnelle de 2 mutations rares de MPL, Tyr591Asn et Ser204Pro, identifiées chez 3 patients TE triples négatifs. Seul un gain de fonction faible a été mis en évidence, suggérant que ces mutations doivent être associées à d’autres anomalies génétiques pour entrainer l’apparition d’un phénotype. / EPOR mutations are observed in Primary familial and congenital polycythaemia (PFCP) while MPL mutations are found in myeloproliferative neoplasms (MPN). PFCP is an inherited disorder of erythroid progenitor cells resulting in elevated erythrocyte mass. Several mutations of the erythropoietin receptor (EPOR) gene have been associated with PFCP. They are all leading to a premature STOP codon and the truncation of the cytoplasmic COOH-terminal of EPOR. To examine the role of EPOR mutations in the pathogenesis of PFCP, we studied a new EPOR mutation, c.1300dupC (p.Gln434Profs*11). This mutation induced, in primary cells and cell lines, a major hypersensitivity to EPO. This phenotype was not due to the loss of negative regulation domains or an internalisation default, contrary to the current hypothesis, but rather due to conformational modification inducing the stabilisation of the mutant at the cell membrane.In the second part of this work, an EPOR mutation, Pro488Ser, was studied in a myeloproliferative neoplasms (MPN) family. Only a mild spontaneous STAT5 activation was observed in cell lines. Murine models and/or iPSC will be developed in order to test the hypothesis of a cooperation between EPOR P488S and JAK2 V617F. In the last part of this project, 2 rare MPL mutants, Tyr591Asn and Ser204Pro, identified in 3 triple negative ET patients were functionally studied. A weak gain of function was observed, suggesting that these mutants have to be associated to other genetic abnormalities to develop a phenotype.
37

Molekulární základy klonální heterogenity hematologických onemocnění / Molecular basis of clonal heterogeneity of hematological diseases

Babošová, Oľga January 2019 (has links)
Tumor heterogeneity has been recognized for decades. The molecular mechanisms impacting clonal heterogeneity in hematological diseases, specifically myeloproliferative neoplasms (MPN) and mantle cell lymphoma, with the focus on several inherited genetic factors, inflammation, the protective mechanisms of DNA damage response (DDR) in the leukemic transformation and the treatment strategies are the focus of this thesis. Firstly, I focus on studying germline JAK2 variants and how these may influence the initiation and progression of MPN diseases, and even contribute to further genomic alterations in the mutated clone. A study performed by our cooperating lab in Utah, USA,1 analyzing the mutational landscape of 31 JAK2 V617F-positive polycythemia vera (PV) patients identified two novel germline mutations in JAK2 gene, JAK2 T108A and JAK2 L393V. Another study2 , performed by our cooperating lab in Olomouc, Czech Republic, characterized two germline JAK2 mutations, E846D and R1063H, in a case of hereditary erythrocytosis accompanied by megakaryocytic atypia. The JAK2 R1063H variant was initially described in 3 out of 93 PV patients that were JAK2 V617F-positive.3 Our aim was to identify the role of selected inherited mutations in JAK2 gene in the initiation and progression of myeloproliferative...
38

Potentiel thérapeutique de l'activation du récepteur nucléaire PPARgamma dans la myélofibrose / Therapeutic Potential of Activation of the Nuclear Receptor PPARgamma Pathway in Myelofibrosis

Lambert, Juliette 16 December 2019 (has links)
La myélofibrose primitive (MFP) est un néoplasme myéloprolifératif (NMP) classique BCR-ABL négatif associé à une forte altération de la qualité de vie et à une augmentation de la mortalité. Les traitements conventionnels réduisent les symptômes mais ont peu d’effet sur l’histoire naturelle de la maladie. La MFP résulte d’interactions complexes entre le développement du clone hématopoïétique malin, l’installation d’un contexte inflammatoire et le remodelage du microenvironnement médullaire. Chacun de ces axes est une cible thérapeutique potentielle. Dans ce travail, nous avons évalué le potentiel thérapeutique de l’activation de PPARγ dans trois modèles murins de myélofibrose et nous montrons que les ligands de PPARγ permettent d’améliorer les paramètres hématologiques et histologiques en rapport avec l’installation du phénotype de myélofibrose. Chacun des axes de la physiopathologie a ensuite été exploré. Les ligands de PPARγ ont une action anti-proliférative sur le clone malin, tant dans les modèles murins de NMPs que dans les cellules JAK2V617F de lignée et dans les progéniteurs hématopoïétiques issus de patients atteints de NMPs. Le traitement atténue également l’hyperleucocytose associée au phénotype inflammatoire des NMPs et modifie la transcription de gènes de l’inflammation. Enfin, les ligands de PPARγ ont un effet protecteur sur le stroma médullaire, dépendant de la capacité de PPARγ à contrecarrer la voie de signalisation du TGF-β1, cytokine majeure du développement de la fibrose médullaire, par déplacement du cofacteur de transcription p300 de la voie du TGF-β1 vers la voie PPARγ. Par son action sur les trois composantes de la physiopathologie, l’activation de PPARγ constitue une cible thérapeutique pertinente dans la prise en charge de la MFP. / Primary myelofibrosis (PMF) is a non BCR-ABL myeloproliferative neoplasm (MPN) associated with poor quality of life and reduced survival. Current treatments are mainly symptomatic and have little effect on the natural history of the disease. PMF results from complex interactions between the emergence of a hematopoietic malignant clone, an inflammatory context and the remodeling of the bone marrow (BM) microenvironment. Each of these axes is a potential therapeutic target. Here, we evaluated the therapeutic potential of PPARγ ligands in three murine models of myelofibrosis and we showed that PPARγ ligands improve hematological and histological changes related to myelofibrosis phenotype. Then, we explored each axis of the pathophysiology. We showed that PPARγ ligands have an anti-proliferative effect and limit the proliferation of the malignant clone in murine models of MPNs, in JAK2V617F cell lines and in hematopoietic progenitors from MPNs patients. PPARγ ligands also decrease leukocytosis related to the inflammatory phenotype of MPNs and modify the transcription of inflammatory genes. Finally, we demonstrated that PPARγ ligands have a protective effect on BM stroma. They counteract the signaling pathway of TGF-β1, a major cytokine in BM fibrosis development, by moving the p300 cofactor of transcription from the TGF-β1 pathway to the PPARγ pathway. By its action on the three components of the pathophysiology, activation of PPARγ pathway is a relevant therapeutic target in PMF.
39

Comparing dropout regularization algorithms for convolutional neural networks identifying malignant cells for diagnosis of leukemia

Engström, Hampus, Koutakis, Alexander January 2023 (has links)
Fast and high quality classifications of cells inflicted with malignant mutations is essential for diagnosing patients with different forms of leukemia, to quickly be able give patients the crucial care they need. Convolutional neural networks (CNNs) can be trained and used for this purpose. This thesis studies CNNs and the application of regularization to create better performing and generalised models, with the purpose of generating highly accurate classifications for nine different forms of malignant white blood cells from the myeloid lineage. This is done to asses what method of dropout regularization is best suited for this type of cell data. To achieve this, three different methods of dropout regularization were studied: Bernoulli dropout; Gaussian dropout; and spatial dropout. This was conducted using a dataset consisting of 106,472 images from 945 patients. The results indicate that models using Gaussian dropout and Bernoulli dropout, respectively, produce the best results, with 87.39\% being the highest accuracy achieved. These two models are also statistically different from a benchmark model not utilizing any form of dropout. This suggests that one of these techniques may be optimal for this type of data. Further studies may be needed to determine which is the best of the two.
40

Linking energy sensing to suppression of JAK-STAT signalling: a potential route for repurposing AMPK activators?

Speirs, C., Williams, Jamie J.L., Riches-Suman, Kirsten, Salt, I.P., Palmer, Timothy M. 13 October 2017 (has links)
Yes / Exaggerated Janus kinase-signal transducer and activator of transcription (JAKSTAT) signalling is key to the pathogenesis of pro-inflammatory disorders, such as rheumatoid arthritis and cardiovascular diseases. Mutational activation of JAKs is also responsible for several haematological malignancies, including myeloproliferative neoplasms and acute lymphoblastic leukaemia. Accumulating evidence links adenosine 5′-monophosphate (AMP)–activated protein kinase (AMPK), an energy sensor and regulator of organismal and cellular metabolism, with the suppression of immune and inflammatory processes. Recent studies have shown that activation of AMPK can limit JAK-STAT-dependent signalling pathways via several mechanisms. These novel findings support AMPK activation as a strategy for management of an array of disorders characterised by hyper-activation of the JAKSTAT pathway. This review discusses the pivotal role of JAK-STAT signalling in a range of disorders and how both established clinically used and novel AMPK activators might be used to treat these conditions. / British Heart Foundation; Diabetes UK; Chief Scientist Office; NHS Greater Glasgow and Clyde Research Endowment Fund; Chest, Heart and Stroke Scotland

Page generated in 0.1164 seconds