• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 5
  • 2
  • 2
  • Tagged with
  • 9
  • 9
  • 6
  • 5
  • 5
  • 5
  • 5
  • 5
  • 5
  • 5
  • 5
  • 4
  • 4
  • 3
  • 3
  • 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

The role of growth differentiation factor 15 in the pathogenesis of primary myelofibrosis / 原発性骨髄線維症の病態におけるGrowth differentiation factor 15の役割

Uchiyama, Tatsuki 23 March 2016 (has links)
京都大学 / 0048 / 新制・課程博士 / 博士(医学) / 甲第19569号 / 医博第4076号 / 新制||医||1017(附属図書館) / 32605 / 京都大学大学院医学研究科医学専攻 / (主査)教授 江藤 浩之, 教授 武藤 学, 教授 中畑 龍俊 / 学位規則第4条第1項該当 / Doctor of Medical Science / Kyoto University / DFAM
2

Impacto da análise molecular da mutação JAK2V617F no diagnóstico de neoplasias mieloproliferativas crônicas de acordo com os critérios da OMS 2016

Pedrazzani, Fabiane Spagnol January 2016 (has links)
As neoplasias mieloproliferativas (NMPs) são um grupo de doenças derivadas de uma transformação clonal de célula tronco hematopoiéticas no qual a linhagem celular mielóide é predominantemente expandida no sangue periférico. As NMPs Philadelphia-negativas incluem policitemia vera (PV), trombocitemia essencial (TE) e mielofibrose primária (MFP) que compartilham muitas características hematológicas, clínicas e evolutivas. A mutação da JAK2 (JAK2V617F) está presente em cerca de 95% dos pacientes com PV, entre 50 a 70% com TE e 40 a 50% com MFP. No entanto, os testes moleculares para diagnóstico são muitas vezes um desafio devido ao alto custo e a disponibilidade de equipamentos especializados. Objetivo: Verificar o impacto do teste molecular da mutação JAK2V617F para o diagnóstico de NMPs nos pacientes atendidos no Hospital de Clínicas de Porto Alegre. Métodos: Foram avaliados 87 pacientes com suspeita de NMPs. As amostras de sangue periférico foram analisadas para a mutação JAK2V617F pelo método genético molecular de PCR alelo-específico e os resultados correlacionados com os dados clínico-laboratoriais. Para estabelecimento do diagnóstico, foram utilizados os critérios da Organização Mundial da Saúde (OMS) de 2016. Resultados: Dos 87 pacientes avaliados, 27,6% foram diagnosticados como PV, 39,1% como TE, 4,6% como MFP e 28,7% não contemplavam os critérios para o diagnóstico NMPs. A comparação da utilização do teste da mutação JAK2V617F mostrou que, apenas 41,7% dos pacientes com PV sem utilizar o teste, teriam sido diagnosticados comparados a 91,7% utilizando este teste como um dos critérios no diagnóstico final (p = 0,004). Na TE e na MFP, este critério não foi estatisticamente significativo. Conclusão: O teste molecular para a mutação de JAK2V617F no nosso hospital teve um impacto significativo no diagnóstico dos pacientes com PV, mostrando ser uma ferramenta importante para o diagnóstico final desta NMP. / Myeloproliferative neoplasms (MPNs) are a group of disorders derived from a clonal transformation of stem cell on which myeloid cell lineage is predominantly expanded in the peripheral blood. Philadelphia-negative MPNs include polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF) which share many hematological, clinical, and evolutionary characteristics. The JAK2 mutation (JAK2V617F) is present in about 95% of patients with PV, between 50 to 70% with ET and 40 to 50% PMF. However, the molecular diagnostic tests are often a challenge due to the high cost and the availability of specialized equipment. Objective: To verify the impact of molecular testing of the JAK2V617F mutation for the diagnosis of MPNs in patients attended at Hospital de Clinics, Porto Alegre. Methods: A total of 97 patients were evaluated with suspected of MPNs. The peripheral blood samples were analyzed for the JAK2V617F mutation by the molecular genetic allelespecific PCR method and the results correlated with the clinical-laboratory data. To establish the diagnosis, the 2016 World Health Organization (WHO) criteria were used. Results: Of the 87 patients evaluated, 27.6% were diagnosed as PV, 39.1% as ET, 4.6% as PMF and 28.7% did not meet criteria for MPNs diagnosis. Comparison of the use of the JAK2V617F test showed that only 41.7% of patients with PV without the mutation test were diagnosed compared to 91.7% using this test as one of the criteria for the final diagnosis (p = 0.004). In the ET and the PMF, this criterion was not statistically significant. Conclusion: The molecular test for the JAK2V617F mutation in our hospital had a significant impact in the diagnosis of patients with PV, showing to be an important tool for the final diagnosis of this MPN.
3

Rôle du couple Flt3-ligand/Flt3 et de l'activation des "Mitogen-activated protein kinases" p38 dans la dysmégacaryopoïèse des patients atteints de myélofibrose primitive / Rôle du couple Flt3-ligand/Flt3 et de l'activation des "Mitogen-activated protein kinases" p38 dans la dysmégacaryopoïèse des patients atteints de myélofibrose primitive

Desterke, Christophe 25 May 2011 (has links)
La myélofibrose primitive (MFP) est un néoplasme myéloprolifératif (NMP) chronique BCR-ABL1-négatif associant une dérégulation de l’hématopoïèse (myéloprolifération, dysmégacaryopoïèse et migration des cellules souches et progéniteurs hématopoïétiques (CSH/PH)) à une altération du stroma médullaire et splénique (fibrose ostéomyélosclérose, néoangiogenèse). Le mégacaryocyte (MK) est un acteur majeur de sa pathogenèse, via la production de cytokines et facteurs fibrosants, dans un contexte inflammatoire. Plusieurs arguments suggèrent que les mutations JAK2V617F et MPL515L/K qui caractérisent les NMP ne sont pas les événements initiaux de la MFP car elles ne sont retrouvées que chez la moitié des patients. L’objectif de mon travail a été de rechercher si d’autres anomalies, géniques ou non, pouvaient expliquer la pathogenèse de la MFP. Pour cela, parallèlement à une démarche génomique (transcriptome et CGH array), nous avons développé une approche de biologie cellulaire ciblée sur le rôle du stroma hématopoïétique. Bien que n’ayant pas identifié d’autres anomalies génomiques que celles décrites dans la littérature et en particulier, la délétion 13q, les approches génomiques que nous avons développées nous ont permis de préciser les bornes de cette délétion dans les PH CD34+ et les polynucléaires des patients. Cette délétion (région chromosomique minimale 13q14-13q21) est située à 2 mégabases (télomérique) du cluster FLT où est localisé le gène FLT3. Plusieurs arguments nous ont ensuite conduits à rechercher si le couple Flt3-ligand/Flt3 était impliqué dans la dérégulation de l’hématopoïèse et plus particulièrement dans la dysmégacaryopoïèse observée chez les patients. Parmi ceux-ci, citons : 1) l’existence d’une modulation d’expression de gènes inclus dans la zone de délétion 13q et dans le cluster FLT, dont le gène FLT3 et 2) le fait que Flt3, un récepteur clé de la régulation de l’hématopoïèse primitive, soit souvent impliqué dans la pathogenèse d’hémopathies malignes et que son ligand, Flt3-ligand, soit majoritairement produit par le stroma hématopoïétique. Notre étude montre une dérégulation de Flt3 et des MAPKs p38 dans les PH CD34+ et les MK des patients atteints de MFP et ceci, quelque soit leur statut mutationnel Jak2. Elle démontre également que la persistance de la stimulation de l’axe Flt3/p38 en réponse à une production accrue de Flt3 ligand, participe à la dysmégacaryopoïèse qui caractérise la maladie. En effet, nous avons mis en évidence : 1) une augmentation du taux sérique de Flt3 ligand et de son expression par les cellules du stroma médullaire et splénique ainsi que par les PH des patients atteints de MFP, 2) une surexpression spécifique de son récepteur Flt3 et de sa phosphorylation dans les CSH/PH CD34+ et les progéniteurs mégacaryocytaires (MK), qui persistent au cours de la différenciation MK, quelque soit le statut mutationnel de Jak2 des patients, 3) une activation de Flt3 dans les progéniteurs MK en réponse au Flt3 ligand conduisant à la phosphorylation en cascade de la voie de signalisation des MAPKs p38 et à l’expression de ses gènes cibles tels que AP-1, p53, NFATc4, ATF2, IL-8, 4) une restauration de la mégacaryopoïèse et une inhibition de la migration (Flt3-ligand)-dépendante des progéniteurs MK des patients après inhibition de Flt3 ou de p38.Nos résultats confirment l’importance d’une altération des MAPKs dans une dérégulation de l’hématopoïèse et soulignent le rôle d’une activation persistante de la voie p38, via le couple Flt3-ligand/Flt3, dans la dysmégacaryopoïèse qui caractérise la myélofibrose primitive. Ils suggèrent également que cette dérégulation participe au processus inflammatoire à l’origine de la réaction stromale et « lit » d’une transformation leucémique potentielle. Ce dialogue altéré entre les cellules hématopoïétiques pathologiques (Bad seeds), en particulier mégacaryocytaires et les cellules stromales (Bad soil), conforte notre concept « Bad seeds in Bad soil ». / The primary myelofibrosis (PMF) is a chronic myeloproliferative neoplasm (NMP) BCR-ABL1-negative associating a dysregulation of hematopoiesis (myeloproliferation, dysmegacaryopoiesis and egress of hematopoietic stem and progenitor cells (HSC / PH)) from an altered bone marrow stroma (osteosclerosis, fibrosis, angiogenesis) to the spleen. The megakaryocyte (MK) is a major player in its pathogenesis through the production of cytokines and fibrotic factors in an inflammatory context. Several arguments suggest that mutations JAK2V617F and MPL515L / K which characterize the NMP are not the initial events of the PMF since they are found only in half of patients. The aim of my work was to investigate whether other abnormalities, genetic or otherwise, could explain the pathogenesis of the PMF. For this, a process parallel to genomics (transcriptome and CGH array), we developed a cell biology approach focused on the role of hematopoietic stroma.Although we have not identified other genomic abnormalities as those described in the literature and in particular, deletion 13q, by genomic approaches we have clarified the limits of this deletion in the PH CD34+ and polymorphonuclear patients. This deletion (chromosomal region 13q14-13q21 minimum) is located 2 megabases (telomeric) of the cluster where is located the FLT gene FLT3. Several arguments have then led to inquire whether the couple was involved in Flt3-ligand/Flt3 deregulation of hematopoiesis, especially in the dysmegakaryopoiesis observed in patients. Among these are: 1) the existence of an expression modulation of genes included in the area of deletion 13q and FLT in the cluster, as gene FLT3 and 2) the fact that Flt3, a key receptor the regulation of primitive hematopoiesis, is often implicated in the pathogenesis of hematologic malignancies and its ligand, Flt3-ligand, was predominantly produced by the hematopoietic stroma.Our study shows dysregulation of Flt3 and p38 MAPKs in CD34+ and PH MK from patients with PMF and this, whatever their Jak2 mutation status. It also shows that persistent stimulation of the axis Flt3/p38 in response to increased production of Flt3 ligand, participates in the dysmegacaryopoiesis that characterizes the disease. Indeed, we have highlighted: 1) an increase in serum Flt3 ligand and its expression by stromal cells and bone marrow and spleen by PH patients with PMF, 2) a specific overexpression of its receptor Flt3 and its phosphorylation in HSC / PH CD34+ and megakaryocytic progenitors (MK), which persist during the MK differentiation, regardless of the mutational status of Jak2 patients, 3) activation of Flt3 in MK progenitors by the Flt3 ligand leads to phosphorylation cascade signaling pathway, p38 MAPK and expression of its target genes such as AP-1, p53, NFATc4, ATF2, IL-8, 4) a restoration of megakaryopoiesis and inhibition of migration (Flt3-ligand)-dependent patients after of MK progenitors by Flt3 or p38 inhibitors.Our results confirm the importance of an alteration of MAPKs in a deregulation of hematopoiesis and highlight the role of a persistent activation of the p38 pathway, via the couple Flt3-ligand/Flt3 in the dysmegakaryopoiesis that characterizes idiopathic myelofibrosis. They also suggest that this dysregulation contributes to the inflammatory process at the origin of the stromal reaction and "bed" of a leukemic transformation potential. The dialogue among impaired hematopoietic cell disease (Bad Seeds), especially the stromal cells and megakaryocyte (Bad Soil), reinforces our concept of "Bad Seeds in Bad Soil". This work could help improve the dialogue with therapeutic approaches targeting the axis Flt3-ligand/Flt3 mediated by activation of p38 which, by reducing the inflammatory process, re-establish a link between the "seed" and the "Soil".
4

Impacto da análise molecular da mutação JAK2V617F no diagnóstico de neoplasias mieloproliferativas crônicas de acordo com os critérios da OMS 2016

Pedrazzani, Fabiane Spagnol January 2016 (has links)
As neoplasias mieloproliferativas (NMPs) são um grupo de doenças derivadas de uma transformação clonal de célula tronco hematopoiéticas no qual a linhagem celular mielóide é predominantemente expandida no sangue periférico. As NMPs Philadelphia-negativas incluem policitemia vera (PV), trombocitemia essencial (TE) e mielofibrose primária (MFP) que compartilham muitas características hematológicas, clínicas e evolutivas. A mutação da JAK2 (JAK2V617F) está presente em cerca de 95% dos pacientes com PV, entre 50 a 70% com TE e 40 a 50% com MFP. No entanto, os testes moleculares para diagnóstico são muitas vezes um desafio devido ao alto custo e a disponibilidade de equipamentos especializados. Objetivo: Verificar o impacto do teste molecular da mutação JAK2V617F para o diagnóstico de NMPs nos pacientes atendidos no Hospital de Clínicas de Porto Alegre. Métodos: Foram avaliados 87 pacientes com suspeita de NMPs. As amostras de sangue periférico foram analisadas para a mutação JAK2V617F pelo método genético molecular de PCR alelo-específico e os resultados correlacionados com os dados clínico-laboratoriais. Para estabelecimento do diagnóstico, foram utilizados os critérios da Organização Mundial da Saúde (OMS) de 2016. Resultados: Dos 87 pacientes avaliados, 27,6% foram diagnosticados como PV, 39,1% como TE, 4,6% como MFP e 28,7% não contemplavam os critérios para o diagnóstico NMPs. A comparação da utilização do teste da mutação JAK2V617F mostrou que, apenas 41,7% dos pacientes com PV sem utilizar o teste, teriam sido diagnosticados comparados a 91,7% utilizando este teste como um dos critérios no diagnóstico final (p = 0,004). Na TE e na MFP, este critério não foi estatisticamente significativo. Conclusão: O teste molecular para a mutação de JAK2V617F no nosso hospital teve um impacto significativo no diagnóstico dos pacientes com PV, mostrando ser uma ferramenta importante para o diagnóstico final desta NMP. / Myeloproliferative neoplasms (MPNs) are a group of disorders derived from a clonal transformation of stem cell on which myeloid cell lineage is predominantly expanded in the peripheral blood. Philadelphia-negative MPNs include polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF) which share many hematological, clinical, and evolutionary characteristics. The JAK2 mutation (JAK2V617F) is present in about 95% of patients with PV, between 50 to 70% with ET and 40 to 50% PMF. However, the molecular diagnostic tests are often a challenge due to the high cost and the availability of specialized equipment. Objective: To verify the impact of molecular testing of the JAK2V617F mutation for the diagnosis of MPNs in patients attended at Hospital de Clinics, Porto Alegre. Methods: A total of 97 patients were evaluated with suspected of MPNs. The peripheral blood samples were analyzed for the JAK2V617F mutation by the molecular genetic allelespecific PCR method and the results correlated with the clinical-laboratory data. To establish the diagnosis, the 2016 World Health Organization (WHO) criteria were used. Results: Of the 87 patients evaluated, 27.6% were diagnosed as PV, 39.1% as ET, 4.6% as PMF and 28.7% did not meet criteria for MPNs diagnosis. Comparison of the use of the JAK2V617F test showed that only 41.7% of patients with PV without the mutation test were diagnosed compared to 91.7% using this test as one of the criteria for the final diagnosis (p = 0.004). In the ET and the PMF, this criterion was not statistically significant. Conclusion: The molecular test for the JAK2V617F mutation in our hospital had a significant impact in the diagnosis of patients with PV, showing to be an important tool for the final diagnosis of this MPN.
5

Impacto da análise molecular da mutação JAK2V617F no diagnóstico de neoplasias mieloproliferativas crônicas de acordo com os critérios da OMS 2016

Pedrazzani, Fabiane Spagnol January 2016 (has links)
As neoplasias mieloproliferativas (NMPs) são um grupo de doenças derivadas de uma transformação clonal de célula tronco hematopoiéticas no qual a linhagem celular mielóide é predominantemente expandida no sangue periférico. As NMPs Philadelphia-negativas incluem policitemia vera (PV), trombocitemia essencial (TE) e mielofibrose primária (MFP) que compartilham muitas características hematológicas, clínicas e evolutivas. A mutação da JAK2 (JAK2V617F) está presente em cerca de 95% dos pacientes com PV, entre 50 a 70% com TE e 40 a 50% com MFP. No entanto, os testes moleculares para diagnóstico são muitas vezes um desafio devido ao alto custo e a disponibilidade de equipamentos especializados. Objetivo: Verificar o impacto do teste molecular da mutação JAK2V617F para o diagnóstico de NMPs nos pacientes atendidos no Hospital de Clínicas de Porto Alegre. Métodos: Foram avaliados 87 pacientes com suspeita de NMPs. As amostras de sangue periférico foram analisadas para a mutação JAK2V617F pelo método genético molecular de PCR alelo-específico e os resultados correlacionados com os dados clínico-laboratoriais. Para estabelecimento do diagnóstico, foram utilizados os critérios da Organização Mundial da Saúde (OMS) de 2016. Resultados: Dos 87 pacientes avaliados, 27,6% foram diagnosticados como PV, 39,1% como TE, 4,6% como MFP e 28,7% não contemplavam os critérios para o diagnóstico NMPs. A comparação da utilização do teste da mutação JAK2V617F mostrou que, apenas 41,7% dos pacientes com PV sem utilizar o teste, teriam sido diagnosticados comparados a 91,7% utilizando este teste como um dos critérios no diagnóstico final (p = 0,004). Na TE e na MFP, este critério não foi estatisticamente significativo. Conclusão: O teste molecular para a mutação de JAK2V617F no nosso hospital teve um impacto significativo no diagnóstico dos pacientes com PV, mostrando ser uma ferramenta importante para o diagnóstico final desta NMP. / Myeloproliferative neoplasms (MPNs) are a group of disorders derived from a clonal transformation of stem cell on which myeloid cell lineage is predominantly expanded in the peripheral blood. Philadelphia-negative MPNs include polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF) which share many hematological, clinical, and evolutionary characteristics. The JAK2 mutation (JAK2V617F) is present in about 95% of patients with PV, between 50 to 70% with ET and 40 to 50% PMF. However, the molecular diagnostic tests are often a challenge due to the high cost and the availability of specialized equipment. Objective: To verify the impact of molecular testing of the JAK2V617F mutation for the diagnosis of MPNs in patients attended at Hospital de Clinics, Porto Alegre. Methods: A total of 97 patients were evaluated with suspected of MPNs. The peripheral blood samples were analyzed for the JAK2V617F mutation by the molecular genetic allelespecific PCR method and the results correlated with the clinical-laboratory data. To establish the diagnosis, the 2016 World Health Organization (WHO) criteria were used. Results: Of the 87 patients evaluated, 27.6% were diagnosed as PV, 39.1% as ET, 4.6% as PMF and 28.7% did not meet criteria for MPNs diagnosis. Comparison of the use of the JAK2V617F test showed that only 41.7% of patients with PV without the mutation test were diagnosed compared to 91.7% using this test as one of the criteria for the final diagnosis (p = 0.004). In the ET and the PMF, this criterion was not statistically significant. Conclusion: The molecular test for the JAK2V617F mutation in our hospital had a significant impact in the diagnosis of patients with PV, showing to be an important tool for the final diagnosis of this MPN.
6

Role of group 2 innate lymphoid cells in the pathogenesis of bone marrow fibrosis / Roll av medfödda lymfoida celler i grupp 2 i patogenesen av benmärgsfibros

Piñero Garasa, Maria Angeles January 2022 (has links)
Primär myelofibros (PMF) är en typ av myeloproliferativ neoplasm (MPN) som leder till en progressiv och irreversibel benmärgsfibros. En somatisk mutation, Jak2V617F, har hittats hos 50 % av patienterna med MPN i hematopoetiska stamceller. Nyligen har man upptäckt grupp 2 av medfödda lymfoida celler (ILC2) som tillhör det medfödda systemet. De är T-cellernas motsvarighet men saknar TCR-receptorn. ILC2 reagerar på IL-33 och producerar Il-13. Under de senaste åren har man upptäckt att dessa två cytokiner är inblandade i PMF. För att undersöka ILC2:s roll i utvecklingen av benmärgsfibros in vivo producerade vi retrovirus som uttrycker Jak2 vildtyp (JAK2_WT) eller Jak2V617F (JAK2_V617F) och transducerade benmärg vildtyp (BM_WT) eller benmärg ILC2KO (BM_ILC2KO). Benmärgen transplanterades till subletalt bestrålade immunbristande möss (NOG). Klinikopatologiska drag som är karakteristiska för sjukdomens första stadier, som förhöjda hemoglobinnivåer, megakaryocythyperplasi och betydande trombocytos, uppstod inte under studieperioden. Ökade vita blodkroppar uppstod dock på grund av avsaknaden av ILC2 i JAK2_V617F-expressiva möss. Flödescytometeranalys visade ursprunget till den markerade leukocytosen som ett resultat av expansionen från lymfocytlinjen, mer specifikt B-celler, men resultaten är inte entydiga eftersom de förhöjda nivåerna av B-celler kan vara en följd av ILC2 knock-out fenotypen som förvärras av närvaron av mutationen. Granulocytnivåerna från de inympade cellerna hölls låga till följd av att stamcellerna i värdens benmärg var inblandade på grund av subletal bestrålning. Vi drar slutsatsen att frånvaron av ILC2 i JAK2_V617F-uttryckta benmärgsprogenitorer har en tendens att förvärra den myeloproliferativa fenotypen i sjukdomens tidiga skeden, vilket tyder på en möjlig skyddande roll för ILC2 vid utvecklingen av MPN. / Primary myelofibrosis (PMF) is one type of myeloproliferative neoplasm (MPN) that leads to a progressive and irreversible bone marrow fibrosis. A somatic mutation, Jak2V617F has been found in 50% of patients with MPN in hematopoietic stem cells. Group 2 innate lymphoid cells (ILC2) belonging to the innate system has been recently discovered. They are the counter part of T cells but lacking the TCR receptor. ILC2 response to IL-33 producing Il-13. In recent years, the involvement of these two cytokines in the PMF has been uncovered. To investigate the role of ILC2 in the progression of bone marrow fibrosis in vivo we produced retrovirus expressing Jak2 wild-type (JAK2_WT) or Jak2V617F (JAK2_V617F) and transduced bone marrow wild type (BM_WT) or bone marrow ILC2KO (BM_ILC2KO). The bone marrow was transplanted into sub-lethally irradiated immunodeficient mice (NOG). Clinicopathologic features characteristic from the first stages of the disease, as elevated hemoglobin levels, megakaryocyte hyperplasia and significant thrombocytosis did not emerge during the study period. However, increased in white blood cells arise from the absence of ILC2 in JAK2_V617F expressing mice. Flow cytometer analysis revealed the origin of the marked leukocytosis as a result of the expansion from the lymphocyte lineage, more specifically B cells, but the results are inconclusive as the elevated levels of B-cells could be a consequence of the ILC2 knock-out phenotype aggravated by the presence of the mutation. Granulocyte levels from engrafted cells were kept low because of the involvement of host bone marrow stem cells due to sublethal irradiation. We conclude that the absence of ILC2 in JAK2_V617F-express bone marrow progenitors has a tendency to aggravate the myeloproliferative phenotype in the early stages of the disease, indicating a possible protective role of ILC2 in the development of MPNs.
7

Anomalies moléculaires et fonctionnelles des cellules stromales mésenchymateuses de patients atteints de myélofibrose primitive : altérations « intrinsèques » de leur différenciation ostéoblastique / Molecular and Functionnal Abnormalities of Mesenchymal Stromal Cells in Primary Myelofibrosis Patients : « intrinsic » Impairment of their Osteogenic Potency

Martinaud, Christophe 18 December 2014 (has links)
La myélofibrose primitive (MFP) est un néoplasme myéloprolifératif chromosome Philadelphie négatif rare, mais de pronostic sévère. Elle se caractérise par une prolifération clonale et une mobilisation des cellules souches et progéniteurs hématopoïétiques (CSH/PH) de la moelle osseuse vers la rate et le foie. Cette anomalie de l’hématopoïèse est associée à une pathologie du stroma (myélofibrose, ostéosclérose et néoangiogenèse). L’existence d’anomalies moléculaires de la CSH/PH telles que les mutations de Jak2, Mpl, TET2 ou CALR ne permet pas à elle seule d’expliquer la physiopathologie de la maladie. Les résultats obtenus dans le laboratoire suggèrent que le microenvironnement médullaire au sein des niches hématopoïétiques et en particulier les cellules stromales mésenchymateuses (CSM), participe vraisemblablement à cette dérégulation de l’hématopoïèse, favorisant le développement du clone pathologique. Cependant, aucune preuve tangible d’une altération des CSM médullaires n’a été jusqu’à présent apportée.Dans ce travail, nous avons isolé les CSM de la moelle de patients atteints de MFP et réalisé une caractérisation « complète » de ces cellules : prolifération, phénotype, soutien de l’hématopoïèse, sécrétome, transcriptome, miRNome et capacités de différenciation. Nos résultats ont permis de dégager un faisceau d’arguments en faveur d’une dérégulation de leur différenciation ostéoblastique (DOB). (i) Les cytokines BMP2, RANTES, PDGF, TGF-β1, VEGF et Il-6 sont significativement produites en plus grande quantité par ces cellules. (ii) L’étude du transcriptome a révélé une expression significativement différente d’un ensemble de gènes impliqués dans la DOB tels que RUNX2, DLX5, TWIST1 et NOGGIN. (iii) De nombreux micro-ARN, dont certains sont connus pour être impliqués dans la DOB comme miR-210 ou dans le nichage des cellules souches hématopoïétiques comme miR-34a, sont dérégulés à l’état basal et au cours de cette DOB. (iv) Enfin, l’étude de leurs capacités de différenciation ostéoblastique in vitro et in vivo chez la souris immunodéprimée est en faveur d’une augmentation de ces capacités. Nous avons étudié l’impact du TGF- β1 dans cette DOB. Nous avons mis en évidence que les CSM de malades présentent un état basal d'activation de la voie de signalisation pSmad significativement augmenté, confirmant l’expression endogène de TGF-β1. En utilisant des inhibiteurs spécifiques du récepteur de type I au TGF- β, nous avons montré l’implication de cette cytokine dans les altérations de la DOB. En conclusion, notre travail montre pour la première fois que les CSM des malades de MFP sont anormales et ce indépendamment de la stimulation par le clone hématopoïétique pathologique, suggérant la présence d'anomalies constitutives ou acquises. Ces anomalies impliquent deux acteurs majeurs de la pathologie : le TGF-β1 et l'ostéogenèse. / Primary myelofibrosis (PMF) is a Philadelphia-negative myeloproliferative neoplasm, rare but associated with a poor prognosis. Its features are a clonal proliferation and an egress of hematopoietic stem cells (HSC) from bone marrow to spleen. These abnormalities of hematopoiesis are in relation with a pathological stroma (myelofibrosis, osteosclerosis and neoangiogenesis). Molecular abnormalities present in HSC partially explain the physiopathology of the disease. Results from our lab suggest that the bone marrow micro-environnement, especially mesenchymal stromal cells (MSC), are involved in the deregulation of hematopoiesis, promoting the clonal cells. However, there is no strong evidence of bone marrow MSC alterations reported for now.In our study, we isolated MSC from bone marrow of patients suffering from PMF and performed a broad characterization: proliferation, phenotype, hematopoiesis supporting capacities, secretome, transcriptome and miRNome analysis. Our results highlight arguments in favor of a deregulation of their osteogenic capacities. (i) Cytokines NMP2, RANTES, PDGF, TGF-β1, VEGF and Il-6 were significantly overproduced by MSCs. (ii) Transcriptome analysis revealed a specific signature involving genes participating in osteogenic differentiation such as RUNX2, DLX5, TWIST1 and NOGGIN. (iii) Many micro-RNAs, some know to be involved in osteogenic differentiation regulation, as mir-34a, are deregulated in MSCs and in MSC-derived osteoblasts. (iv) Finally, study of their osteogenic potency in vitro and in vivo in nude mice showed an increasing of their osteogenic potency. We studied the impact of TGF-β1 in this process and showed that PMF MSCs showed a basal expression of Smad pathway significantly increased as compared to control. Using specific inhibitor of TGF-β1 receptor, we demonstrated the implication of this cytokine in the osteogenic impairment.To summarize, our work shows for the first time that MSCs from PMF patients are abnormal, independently from stimulation by clonal cells, suggesting intrinsic abnormalities. These abnormalities involve two main factor of the disease: TGF-β1 and osteogenesis.
8

Estudo do perfil genético de pacientes com Neoplasias Mieloproliferativas (NMP) cromossomo Filadélfia negativo / Study of genetic profile of patients with Philadelphia-negative Myeloproliferative Neoplasms (MPN)

Marchiani, Mariana 26 February 2016 (has links)
As neoplasias mieloproliferativas (NMPs) Filadelfia negativo como a policitemia vera (PV), trombocitemia essencial (TE) e mielofibrose primária (MFP) são desordens clonais da célula tronco hematopoiética caracterizadas pela produção excessiva de células mielóides diferenciadas. Este fenômeno ocorre devido à uma mutação somática (JAK2V617F) que ativa a via JAK-STAT de transdução de forma constitutiva. Esta mutação é mais frequente na PV, ocorrendo em 95% dos casos, e em 50% dos casos de TE e MFP. Outro defeito genético que ocorre é a mutação no receptor de trombopoetina, MPL. As mutações em MPL podem ser germinativas ou somáticas e menos de 10% dos pacientes com TE e MFP apresentam essa alteração genética. Entretanto, grande parte dos pacientes com TE e MFP que não apresentam mutação em JAK2V617F ou MPL podem apresentar mutações somáticas no gene CALR. Em adição às mutações somáticas que causam mieloproliferação, outras alterações genéticas em genes que funcionam como reguladores epigenéticos são encontrados nas NMPs nos genes TET2, IDH1, IDH2 e ASLX1. Objetivo: Estabelecer um perfil genético em pacientes com NMP através da avaliação de mutações nos genes JAK2, CALR, MPL, IDH1, IDH2, TET2 e ASXL1 assim como estabelecer uma correlação laboratorial destas na PV, TE e MFP. Casuística e Métodos: Foram utilizadas amostras de sangue periférico de 104 pacientes que foram enviadas para o Laboratório de Biologia Tumoral do Serviço de Hematologia do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo para avaliação diagnóstica. Quinze dos 104 pacientes são de pacientes com PV (14,4%), 20/104 (19,2%) com MFP, 20/104 (19,2%) com TE e 49/104 (47,1%) com outras doenças hemtológicas. Foi feita a avaliação da prevalência de mutações somáticas, seja por sequenciamento ou análise de fragmentos, nos genes JAK2 (exon 12 e Y931C), MPL, IDH1, IDH2, CALR, TET2 e ASXL1. PCR-RFLP foi realizada para identificação de mutações em JAK2V617F. Resultados: A mutação JAK2V617F foi observada em 30 (28,8%) pacientes (12 PV, 11 TE e 7 MFP), a mutação JAK2 exon 12 foi observada em apenas um (0,96%) paciente com PV, mutação JAK2Y931C em 4 (3,8%) pacientes (1 PV, 2 TE e 1 MFP) e 8 (7,7%) pacientes apresentaram mutações em CALR (3 TE e 5 MFP). Mutações nos genes epigenéticos como IDH1 foram observadas em 9 (8,7%) pacientes (2 TE, 2 MFP, 1 SMD, e 4 pacientes com suspeita de NMP), mutações em IDH2 estão presentes em 5 (4,8%) pacientes (2 TE, 1 SMD/leucemia e 4 pacientes com suspeita de NMP), mutações em ASXL1 foram identificadas em 13 (12,5%) pacientes (1 PV, 3 TE, 2 MFP, 3 SMD/leucemias e 4 com suspeita de NMP) e finalmente, mutações em TET2 foram encontradas em 33 (31,7%) pacientes (3 PV, 5 TE, 4 MFP, 8 SMD/leucemias e 13 pacientes com suspeita de NMP). Além disso, no caso da PV, os pacientes que apresentam mutações em JAK2V617F apresentam valores aumentados de plaquetas (mediana de 5,41 x 105/mm3 plaquetas) em relação aos pacientes sem a mutação (mediana de 2,06 x 105/mm3 plaquetas), com diferença estatística (p=0,031). Pacientes do mesmo grupo que apresentam mutações em TET2 apresentam, opostamente aos com mutações em JAK2V617F, menores valores de plaquetas (mediana de 1,75 x105/mm3 plaquetas) em relação aos pacientes sem mutações no gene TET2 (mediana de 5,41 x 105/mm3 plaquetas), com diferença estatística (p=0,048). No caso da MFP, os pacientes que apresentam mutações em JAK2V617F apresentam valores maiores de leucócitos (mediana de 1,09 x104/mm3 leucócitos) do que os pacientes que não apresentam a mutação (mediana de 6,99 x103/mm3 leucócitos) com diferença estatística (p=0,046), já os pacientes que apresentam mutações no gene ASXL1 apresentam valores menores de hemácias (mediana de 2,43 x106/mm3 hemácias) em relação aos pacientes que não apresentam mutação (mediana de 3,71 x106/mm3 hemácias) com diferença estatística (p=0,042). Conclusão: O trabalho permitiu fornecer um perfil genético dos pacientes com NMP estudados. Além disso, é possível observar que algumas mutações epigenéticas podem influenciar em diferenças clínicas / Myeloproliferative neoplasms (MPNs) Philadelphia (Ph) chromosome negative, such as polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF) are clonal disorders of hematopoietic stem cell characterized by increased proliferation of differentiated myeloid cells. This phenomenon occurs due somatic mutation (JAK2V617F) that constitutively stimulates the JAK-STAT signaling pathway. This mutation is more frequent in PV, around 95%, and between 50% in ET and PMF. Other genetic aberration can be observed in the thrombopoietin (TPO) receptor MPL. Mutations in MPL can be in the germline line or somatic and less than 10% of patients with TE or PMF would harbor this genetic alteration. Otherwise, patients with TE or PMF without JAK2V617F or MPL mutation could present somatic mutations in calreticulin (CALR). In addition to somatic mutations that cause myeloproliferation, other genetic alterations that function as epigenetic regulators were identified in genes as TET2, IDH1, IDH2 e ASLX1 in MPN. Objective: Establish genetic profile in patients with diagnosis of PV, ET, and PMF through genetic alterations in the following genes: JAK2, MPL, CALR, IDH1, IDH2, TET2 e ASXL1, and correlate those alterations with demographic characteristic of the study population. Casuistic and Methods: Peripheral blood samples from 104 patients referred to the Tumor Biology Laboratory of the Department of Hematology of Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo for diagnostic investigation were analyzed. Fifteen out 104 samples were from PV patients (14.4%), 20/104 (19.2%) in PMF, 20/104 (19.2%) in ET and 49/104 (47.1%) with other hematologic diseases. Identification of somatic mutations was made, either by direct sequencing or fragment analysis in JAK2 (exon 12 and Y931C), MPL, IDH1, IDH2, CALR, TET2 and ASXL1. PCR-RFLP was performed to identify JAK2V617F mutation. Results: JAK2V617F mutation was observed in 30 (28.8%) patients (12 PV, 11 ET and 7 PMF), JAK2 exon 12 in only one (0.96%) patient with PV, JAK2Y931C in 4 (3.8%) patients (1 PV, 2 ET and 1 PMF), and 8 patients (7.7%) presented CALR mutation (3 ET and 5 PMF). Mutations in the epigenetic genes as IDH1 were observed in 9 (8.7%) patients (2 ET, 2 PMF, 1 MDS and 4 patients with suspected MPN), IDH2 mutations were present in 5 (4.8%) patients (2 ET, 1 MDS/leukemia, and 4 patients with suspected MPN), ASLX1 mutations were identified in 13 (12.5%) patients (1 PV, 3 ET, 2 PMF, 3 MDS/leukemia and 4 with suspected MPN) and finally, TET2 mutations were present in 33 (31.7%) patients (3 PV, 5 ET, 4 PMF, 8 MDS/leukemia, and 13 with suspected MPN). In addition, patients with PV who harbor JAK2V617F have increased platelet counts (median 5.41 x 105/mm3 platelets) compared to those without the mutation (median 2.06 x 105/mm3 platelets, p=0.031). Patients in the same group with TET2 mutation, as opposed to those with JAK2V617F, presented low platelets counts (median of 1.75 x 105/mm3 platelets) compared to those without TET2 mutation (median 5.41 x 105/mm3 platelets, p=0.048). Presence of JAK2V617F in patients diagnosed with PMF have a greater number of leukocytes (median 1.09 x104/mm3 leukocytes) when compared to patients without the mutation (median 6.99 x 103/mm3 leukocytes, p=0.046). Patients with PMF who presented mutations in ASXL1 gene have a lower number of red blood cells (median of 2.43 x 106/mm3) compared to patients without mutations in the same gene (median 3.71 x 106/mm3, p=0.042). Conclusion: The present study allows us to provide a genetic profile of patients with MPN. Furthermore, it is possible to observe that some epigenetic mutations could influence in some clinical differences
9

Estudo do perfil genético de pacientes com Neoplasias Mieloproliferativas (NMP) cromossomo Filadélfia negativo / Study of genetic profile of patients with Philadelphia-negative Myeloproliferative Neoplasms (MPN)

Mariana Marchiani 26 February 2016 (has links)
As neoplasias mieloproliferativas (NMPs) Filadelfia negativo como a policitemia vera (PV), trombocitemia essencial (TE) e mielofibrose primária (MFP) são desordens clonais da célula tronco hematopoiética caracterizadas pela produção excessiva de células mielóides diferenciadas. Este fenômeno ocorre devido à uma mutação somática (JAK2V617F) que ativa a via JAK-STAT de transdução de forma constitutiva. Esta mutação é mais frequente na PV, ocorrendo em 95% dos casos, e em 50% dos casos de TE e MFP. Outro defeito genético que ocorre é a mutação no receptor de trombopoetina, MPL. As mutações em MPL podem ser germinativas ou somáticas e menos de 10% dos pacientes com TE e MFP apresentam essa alteração genética. Entretanto, grande parte dos pacientes com TE e MFP que não apresentam mutação em JAK2V617F ou MPL podem apresentar mutações somáticas no gene CALR. Em adição às mutações somáticas que causam mieloproliferação, outras alterações genéticas em genes que funcionam como reguladores epigenéticos são encontrados nas NMPs nos genes TET2, IDH1, IDH2 e ASLX1. Objetivo: Estabelecer um perfil genético em pacientes com NMP através da avaliação de mutações nos genes JAK2, CALR, MPL, IDH1, IDH2, TET2 e ASXL1 assim como estabelecer uma correlação laboratorial destas na PV, TE e MFP. Casuística e Métodos: Foram utilizadas amostras de sangue periférico de 104 pacientes que foram enviadas para o Laboratório de Biologia Tumoral do Serviço de Hematologia do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo para avaliação diagnóstica. Quinze dos 104 pacientes são de pacientes com PV (14,4%), 20/104 (19,2%) com MFP, 20/104 (19,2%) com TE e 49/104 (47,1%) com outras doenças hemtológicas. Foi feita a avaliação da prevalência de mutações somáticas, seja por sequenciamento ou análise de fragmentos, nos genes JAK2 (exon 12 e Y931C), MPL, IDH1, IDH2, CALR, TET2 e ASXL1. PCR-RFLP foi realizada para identificação de mutações em JAK2V617F. Resultados: A mutação JAK2V617F foi observada em 30 (28,8%) pacientes (12 PV, 11 TE e 7 MFP), a mutação JAK2 exon 12 foi observada em apenas um (0,96%) paciente com PV, mutação JAK2Y931C em 4 (3,8%) pacientes (1 PV, 2 TE e 1 MFP) e 8 (7,7%) pacientes apresentaram mutações em CALR (3 TE e 5 MFP). Mutações nos genes epigenéticos como IDH1 foram observadas em 9 (8,7%) pacientes (2 TE, 2 MFP, 1 SMD, e 4 pacientes com suspeita de NMP), mutações em IDH2 estão presentes em 5 (4,8%) pacientes (2 TE, 1 SMD/leucemia e 4 pacientes com suspeita de NMP), mutações em ASXL1 foram identificadas em 13 (12,5%) pacientes (1 PV, 3 TE, 2 MFP, 3 SMD/leucemias e 4 com suspeita de NMP) e finalmente, mutações em TET2 foram encontradas em 33 (31,7%) pacientes (3 PV, 5 TE, 4 MFP, 8 SMD/leucemias e 13 pacientes com suspeita de NMP). Além disso, no caso da PV, os pacientes que apresentam mutações em JAK2V617F apresentam valores aumentados de plaquetas (mediana de 5,41 x 105/mm3 plaquetas) em relação aos pacientes sem a mutação (mediana de 2,06 x 105/mm3 plaquetas), com diferença estatística (p=0,031). Pacientes do mesmo grupo que apresentam mutações em TET2 apresentam, opostamente aos com mutações em JAK2V617F, menores valores de plaquetas (mediana de 1,75 x105/mm3 plaquetas) em relação aos pacientes sem mutações no gene TET2 (mediana de 5,41 x 105/mm3 plaquetas), com diferença estatística (p=0,048). No caso da MFP, os pacientes que apresentam mutações em JAK2V617F apresentam valores maiores de leucócitos (mediana de 1,09 x104/mm3 leucócitos) do que os pacientes que não apresentam a mutação (mediana de 6,99 x103/mm3 leucócitos) com diferença estatística (p=0,046), já os pacientes que apresentam mutações no gene ASXL1 apresentam valores menores de hemácias (mediana de 2,43 x106/mm3 hemácias) em relação aos pacientes que não apresentam mutação (mediana de 3,71 x106/mm3 hemácias) com diferença estatística (p=0,042). Conclusão: O trabalho permitiu fornecer um perfil genético dos pacientes com NMP estudados. Além disso, é possível observar que algumas mutações epigenéticas podem influenciar em diferenças clínicas / Myeloproliferative neoplasms (MPNs) Philadelphia (Ph) chromosome negative, such as polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF) are clonal disorders of hematopoietic stem cell characterized by increased proliferation of differentiated myeloid cells. This phenomenon occurs due somatic mutation (JAK2V617F) that constitutively stimulates the JAK-STAT signaling pathway. This mutation is more frequent in PV, around 95%, and between 50% in ET and PMF. Other genetic aberration can be observed in the thrombopoietin (TPO) receptor MPL. Mutations in MPL can be in the germline line or somatic and less than 10% of patients with TE or PMF would harbor this genetic alteration. Otherwise, patients with TE or PMF without JAK2V617F or MPL mutation could present somatic mutations in calreticulin (CALR). In addition to somatic mutations that cause myeloproliferation, other genetic alterations that function as epigenetic regulators were identified in genes as TET2, IDH1, IDH2 e ASLX1 in MPN. Objective: Establish genetic profile in patients with diagnosis of PV, ET, and PMF through genetic alterations in the following genes: JAK2, MPL, CALR, IDH1, IDH2, TET2 e ASXL1, and correlate those alterations with demographic characteristic of the study population. Casuistic and Methods: Peripheral blood samples from 104 patients referred to the Tumor Biology Laboratory of the Department of Hematology of Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo for diagnostic investigation were analyzed. Fifteen out 104 samples were from PV patients (14.4%), 20/104 (19.2%) in PMF, 20/104 (19.2%) in ET and 49/104 (47.1%) with other hematologic diseases. Identification of somatic mutations was made, either by direct sequencing or fragment analysis in JAK2 (exon 12 and Y931C), MPL, IDH1, IDH2, CALR, TET2 and ASXL1. PCR-RFLP was performed to identify JAK2V617F mutation. Results: JAK2V617F mutation was observed in 30 (28.8%) patients (12 PV, 11 ET and 7 PMF), JAK2 exon 12 in only one (0.96%) patient with PV, JAK2Y931C in 4 (3.8%) patients (1 PV, 2 ET and 1 PMF), and 8 patients (7.7%) presented CALR mutation (3 ET and 5 PMF). Mutations in the epigenetic genes as IDH1 were observed in 9 (8.7%) patients (2 ET, 2 PMF, 1 MDS and 4 patients with suspected MPN), IDH2 mutations were present in 5 (4.8%) patients (2 ET, 1 MDS/leukemia, and 4 patients with suspected MPN), ASLX1 mutations were identified in 13 (12.5%) patients (1 PV, 3 ET, 2 PMF, 3 MDS/leukemia and 4 with suspected MPN) and finally, TET2 mutations were present in 33 (31.7%) patients (3 PV, 5 ET, 4 PMF, 8 MDS/leukemia, and 13 with suspected MPN). In addition, patients with PV who harbor JAK2V617F have increased platelet counts (median 5.41 x 105/mm3 platelets) compared to those without the mutation (median 2.06 x 105/mm3 platelets, p=0.031). Patients in the same group with TET2 mutation, as opposed to those with JAK2V617F, presented low platelets counts (median of 1.75 x 105/mm3 platelets) compared to those without TET2 mutation (median 5.41 x 105/mm3 platelets, p=0.048). Presence of JAK2V617F in patients diagnosed with PMF have a greater number of leukocytes (median 1.09 x104/mm3 leukocytes) when compared to patients without the mutation (median 6.99 x 103/mm3 leukocytes, p=0.046). Patients with PMF who presented mutations in ASXL1 gene have a lower number of red blood cells (median of 2.43 x 106/mm3) compared to patients without mutations in the same gene (median 3.71 x 106/mm3, p=0.042). Conclusion: The present study allows us to provide a genetic profile of patients with MPN. Furthermore, it is possible to observe that some epigenetic mutations could influence in some clinical differences

Page generated in 0.4494 seconds