• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 11
  • 6
  • 6
  • Tagged with
  • 23
  • 13
  • 11
  • 11
  • 9
  • 9
  • 9
  • 8
  • 7
  • 6
  • 5
  • 5
  • 5
  • 5
  • 4
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
11

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.
12

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.
13

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.
14

Alterações da expressão de apoptomirs, de genes e proteínas pró- e anti-apoptóticos em Mielofibrose e Trombocitemia Essencial / Deregulated expression of apoptomirs and apoptosis-related genes and proteins in Primary Myelofibrosis and Essential Thrombocythemia

Tognon-Ribeiro, Raquel 11 October 2011 (has links)
As Neoplasias Mieloproliferativas Crônicas (NMPC) Trombocitemia Essencial (TE), Mielofibrose (MF) - são desordens hematopoéticas resultantes da expansão clonal da célula tronco hematopoética alterada. Essas doenças caracterizam-se pela independência dos progenitores hematopoéticos aos estímulos dos fatores de crescimento e citocinas e pela proliferação exacerbada das células da linhagem mielóide com maturação preservada. Os mecanismos moleculares e celulares envolvidos na patogênese e progressão da TE e MF não foram ainda esclarecidos, mas o mieloacúmulo presente nessas doenças parece estar associado à alteração de proliferação e apoptose celular. Nesse contexto, os objetivos deste trabalho foram avaliar em células CD34+ da medula óssea e leucócitos dos pacientes com TE e MF: (1) a expressão de apoptomirs e de genes pró- e anti-apoptóticos pertencentes à via intrínseca e extrínseca da apoptose pela metodologia de RT-PCR em tempo real; (2) expressão de proteínas pró- e anti-apoptóticas por Western-blot; (3) o perfil de sensibilidade das células mononucleares à apoptose induzida por diferentes agentes apoptogênicos pela técnica de citometria de fluxo e (4) correlacionar os resultados obtidos com dados clínico-laboratoriais dos pacientes e com a expressão do gene PRV-1. Em TE, nas células CD34+, foi detectado aumento da expressão de a1, mcl-1, bid, bok e noxa e diminuição de bax em relação aos controles. Nos leucócitos, foi detectada a elevação da expressão de a1, bcl-2, bcl-w e bcl-xL, bad e bok e diminuição de bid e bimEL. Em comparação com o grupo controle, os pacientes com MF apresentaram maior expressão dos genes a1, bcl-w bak, bok e noxa e menor de bcl-2 nas células CD34+. Nos leucócitos dos pacientes com MF foi verificado aumento da expressão dos genes bcl-2, bcl-w e bcl-xL, bad e bok. O c-iap-1 apresentou maior expressão nas células CD34+ dos pacientes com MF e TE, enquanto que o c-iap-2 estava elevado nos leucócitos e células CD34+. Foi ainda detectada a expressão diferencial em TE e MF dos genes pertencentes a via de receptor de morte, como a maior expressão dos genes fas, fas-L, faim e dr4 nas células CD34+ dos pacientes e menor expressão do fas-L e trail nos leucócitos dos pacientes com TE e MF. Os resultados indicaram associação da expressão do gene PRV-1 com a mutação JAK2V617F e com a expressão dos genes a1, bcl-w, bik, bax, c-iap-2 e trail. Com relação à expressão proteica, BCL-XL estava aumentada e TRAIL diminuída em leucócitos de pacientes com MF enquanto que a molécula BID estava diminuída em leucócitos de pacientes com TE. Os miRNA26a, let 7d e miR15a estavam mais expressos nos leucócitos de pacientes com TE e MF. Em pacientes com TE o miR130b estava elevado e o mIR16 diminuído, enquanto que nos pacientes com MF o miR198 estava aumentado. Os linfócitos dos pacientes com TE e MF apresentaram maior resistência à apoptose estimulada por: actinomicina, etoposídeo, teniposídeo, citarabina e estaurosporina do que os linfócitos dos controles. Em conclusão, os dados obtidos sugerem a ligação da alteração do processo de apoptose celular, com a expressão do gene PRV-1 e status da mutação JAK2V617F. Esses resultados contribuem para o melhor entendimento da fisiopatologia das NMPC visto que associam a fisiopatologia da TE e MF com a resistência das células alteradas à apoptose. Essas informações serão úteis no futuro, possibilitando o desenho de novos alvos terapêuticos e a descrição de novos marcadores de prognóstico. / Chronic Myeloproliferative Neoplasms (cMPN) - Essential Thrombocythemia (ET), Primary Myelofibrosis (PMF) and Polycythemia Vera (PV) - are clonal hematopoietic stem cell malignancies characterized by an accumulation of mature myeloid cells in bone marrow and peripheral blood. It seems that apoptotic machinery deregulation contribute to ET and PMF pathogenesis. Despite the advances in the molecular knowledge, the physiopathology of these diseases remains unknown. This study investigated cellular and molecular mechanisms involved in apoptosis process regulation in bone marrow haematopoietic progenitor CD34+ cells and leukocytes from ET and PMF patients. The specifics aims were: (1) to evaluate death receptors family members and Bcl-2 related genes expression as well apoptosis-related microRNAs by Real Time PCR; (2) apoptosis-related protein expression by Western Blot; (3) access mononuclear cells apoptosis resistance by flow cytometry and (4) to correlate the results with JAK2V617F mutation, PRV-1 gene expression and as well clinical data. In ET CD34+ cells, we found overexpression of a1, mcl-1, bid, bok e noxa and a decrease of bax compared to controls, while in leukocytes a1, bcl-2, bcl-w e bcl-xL, bad e bok expression was increased and bid and bimEL expression was lower than in controls. In PMF, a1, bcl-w bak, bok e noxa were overexpressed and bcl-2 downregulated in CD34+ cells. In PMF leukocytes bcl-2, bcl-w e bcl-xL, bad e bok mRNA levels were increased. c-iap-1 was increased in ET and PMF CD34+ cells and c-iap-2 expression elevated in ET and PMF CD34+ cells and leukocytes. Death receptor related genes showed overexpression of fas, fas-L, faim and dr4 in patients CD34+ cells and downregulation of fas-L and trail in ET and PMF leukocytes. We found differential expression of several genes between patients JAK2V617F positive and negative, as well we found correlation between gene expression and JAK2V617F allele burden, white blood cells and platelets count and splenomegaly. PRV-1 gene was overexpressed in ET and PMF leukocytes and showed correlation with JAK2V617F mutation and a1, bcl-w, bik, bax, c-iap-2 and trail gene expression. Regarding protein expression, BCL-XL was increased and TRAIL decreased in PMF leukocytes and BID was decreased in ET leukocytes. miRNA26a, let 7d e miR15a was overexpressed in ET and PMF leukocytes, while miR130b was increased only in ET and miR198 only in PMF. miR16 was downregulated in ET leukocytes comparing to controls. We also detected a resistance to apoptosis-inducers in ET and PMF lymphocytes and we observed correlation between apoptosis percentage and the expression of many studied genes. In conclusion, the results indicate the participations of Bcl-2 family genes and Death Receptor pathway genes, as well PRV-1 and JAK2V617F mutation in these disorders, which contribute to elucidate cMPN physiopathology and might lead to the discovery of new cMPN therapies and molecular markers.
15

Severe Pulmonary Hypertension in Chronic Idiopathic Myelofibrosis

Halank, Michael, Marx, C., Baretton, Gustavo B., Müller, K.-M., Ehninger, Gerhard, Höffken, Gerd 24 February 2014 (has links) (PDF)
Background: Chronic myeloproliferative disorders (CMPD) seem to be associated with an increased risk for pulmonary hypertension (PH). Case Report: A patient with history of chronic idiopathic myelofibrosis (CIMF) presented with progressive dyspnea (New York Heart Association class III). Until this time he had not received specific treatment for CIMF. Echocardiography and rightheart catheterization confirmed PH. Further diagnostic procedures excluded a specific cause of PH. Therefore, primary PH was assumed. 2 years later he presented again with progressive dyspnea due to a progress of PH. A few days later the patient died from acute posterior myocardial infarction. Pathologic examination of the lung showed an obstruction of the small vessels by conglomerates of megakaryocytes. Discussion: We conclude that PH developed secondarily due to CMPD. PH should be suspected in patients with CMPD and should influence the decision for treatment of CMPD. / Hintergrund: Chronische myeloproliferative Erkrankungen (CMPD) scheinen mit einem erhöhten Risiko für pulmonale Hypertonie (PH) assoziiert zu sein. Kasuistik: Ein Patient mit chronisch idiopathischer Myelofibrose (CIMF) wurde aufgrund einer progressiven Belastungsdyspnoe (New York Heart Association Stadium III) überwiesen. Bis zu diesem Zeitpunkt erhielt er keine spezifische Behandlung seiner CIMF. Echokardiographie und Rechtsherzkatheter ergaben das Vorliegen einer PH. Eine spezifische Ursache der PH konnte zunächst ausgeschlossen werden. Somit wurde das Vorliegen einer primären PH vermutet. 2 Jahre später wurde der Patient mit erneut verschlechterter Belastungsdyspnoe vorgestellt, wobei ein Progress der PH feststellbar war. Einige Tage später verstarb der Patient an einem Hinterwandinfarkt. Die Autopsie des Lungengewebes zeigte einen Verschluss der kleinen Lungengefäße durch Konglomerate von Megakaryozyten. Diskussion: Die Entwicklung der PH ist bei diesem Patienten als Folge der CMPD einzuschätzen. Das Vorliegen einer PH bei Patienten mit CMPD sollte die Entscheidung zu spezifischen therapeutischen Maßnahmen hinsichtlich der CMPD beeinflussen. / Dieser Beitrag ist mit Zustimmung des Rechteinhabers aufgrund einer (DFG-geförderten) Allianz- bzw. Nationallizenz frei zugänglich.
16

Caractérisation moléculaire des syndromes myéloprolifératifs non leucémie myéloïde chronique / Molecular characterization of myeloproliferative neoplasms non-Chronic Myeloid Leukemia

Brecqueville, Mandy 27 September 2013 (has links)
Les syndromes myéloprolifératifs (SMP) non leucémie myéloïde chronique (LMC) sont des hémopathies myéloïdes chroniques affectant la cellule souche hématopoïétique, pouvant évoluer en leucémie aigüe myéloïde (LAM). Les SMP non LMC incluent la polyglobulie de Vaquez (PV), la thrombocytémie essentielle (TE) et la myélofibrose (MF). La mutation JAK2V617F est retrouvée dans 97% des cas de PV et dans 50% des cas de TE et MF ; elle n'est pas indispensable à la physiopathologie des SMP car JAK2 n'est pas muté à 100%. Afin de progresser dans la compréhension de la physiopathologie des SMP et afin d'identifier de nouveaux marqueurs moléculaires pour le diagnostic, le suivi et le pronostic; nous avons étudié des échantillons de PV, TE, MF ainsi que des LAM post-SMP. Nous avons utilisé des approches moléculaires complémentaires: séquençage, hybridation génomique comparative (CGH-array) et profils d'expression génique. Nous avons identifié des mutations de gènes régulateurs de l'épigénétique (ASXL1, TET2, DNMT3A, SUZ12) et des gènes de la machinerie de l'épissage de l'ARN (SF3B1, SRSF2). Nous avons également identifié que les co-mutations des gènes JAK2 et ASXL1 étaient associées à un mauvais pronostic. Au sein du sous-type MF, nous avons identifié par CGH-array des aberrations du nombre de copies des gènes. Celles-ci contiennent plusieurs gènes candidats susceptibles de participer à la physiopathologie des MF et à l'évolution en LAM (délétion 20q, NF1, ETV6). Nos travaux sur la caractérisation moléculaire des SMP contribuent à l'évolution vers une classification moléculaire avec l'objectif d'une médecine de précision où chaque SMP sera traité en fonction de ses altérations. / Myeloproliferative neoplasms (MPN) are chronic and clonal stem cell myeloid disorders, which can evolve to acute myeloid leukemia (AML). MPN non-chronic myeloid leukemia (CML) include Polycythemia Vera (PV), Essential Thrombocythemia (ET) and Myelofibrosis (MF) (primary or secondary to PV/ET). JAK2V617F mutation is found in 97% of PV and in around half of patients with ET or MF. Nevertheless, this mutation is not essential for MPN physiopathology, because in half of ET/MF cases, JAK2 is not mutated. To progress in the knowledge of MPN physiopathology and in particular of MF; and to find new molecular markers for MPN diagnosis, disease course, and prognosis, we studied several samples of PV, ET, MF and post-AML. We used gene sequencing, array-Comparative Genomic Hybridization (aCGH) and gene expression analyses. We identified several mutations in genes implicated in Epigenetic regulation (ASXL1, TET2, DNMT3A, SUZ12) and in genes implicated in the RNA splicing machinery (SF3B1, SRSF2). We also found that JAK2 and ASXL1 co-mutation is associated with a poor prognosis. In MF, we found by aCGH several copy number aberrations that involve potential leukemogenic genes. Our gene expression data support the hypothesis that PV, ET and MF are a continuum of the same pathology. Our results on molecular characterization help establish a new molecular classification of MPNs with the objective personalized treatment where each MPN will be treated depending on the alterations present in the myeloid cell genome.
17

Alterações da expressão de apoptomirs, de genes e proteínas pró- e anti-apoptóticos em Mielofibrose e Trombocitemia Essencial / Deregulated expression of apoptomirs and apoptosis-related genes and proteins in Primary Myelofibrosis and Essential Thrombocythemia

Raquel Tognon-Ribeiro 11 October 2011 (has links)
As Neoplasias Mieloproliferativas Crônicas (NMPC) Trombocitemia Essencial (TE), Mielofibrose (MF) - são desordens hematopoéticas resultantes da expansão clonal da célula tronco hematopoética alterada. Essas doenças caracterizam-se pela independência dos progenitores hematopoéticos aos estímulos dos fatores de crescimento e citocinas e pela proliferação exacerbada das células da linhagem mielóide com maturação preservada. Os mecanismos moleculares e celulares envolvidos na patogênese e progressão da TE e MF não foram ainda esclarecidos, mas o mieloacúmulo presente nessas doenças parece estar associado à alteração de proliferação e apoptose celular. Nesse contexto, os objetivos deste trabalho foram avaliar em células CD34+ da medula óssea e leucócitos dos pacientes com TE e MF: (1) a expressão de apoptomirs e de genes pró- e anti-apoptóticos pertencentes à via intrínseca e extrínseca da apoptose pela metodologia de RT-PCR em tempo real; (2) expressão de proteínas pró- e anti-apoptóticas por Western-blot; (3) o perfil de sensibilidade das células mononucleares à apoptose induzida por diferentes agentes apoptogênicos pela técnica de citometria de fluxo e (4) correlacionar os resultados obtidos com dados clínico-laboratoriais dos pacientes e com a expressão do gene PRV-1. Em TE, nas células CD34+, foi detectado aumento da expressão de a1, mcl-1, bid, bok e noxa e diminuição de bax em relação aos controles. Nos leucócitos, foi detectada a elevação da expressão de a1, bcl-2, bcl-w e bcl-xL, bad e bok e diminuição de bid e bimEL. Em comparação com o grupo controle, os pacientes com MF apresentaram maior expressão dos genes a1, bcl-w bak, bok e noxa e menor de bcl-2 nas células CD34+. Nos leucócitos dos pacientes com MF foi verificado aumento da expressão dos genes bcl-2, bcl-w e bcl-xL, bad e bok. O c-iap-1 apresentou maior expressão nas células CD34+ dos pacientes com MF e TE, enquanto que o c-iap-2 estava elevado nos leucócitos e células CD34+. Foi ainda detectada a expressão diferencial em TE e MF dos genes pertencentes a via de receptor de morte, como a maior expressão dos genes fas, fas-L, faim e dr4 nas células CD34+ dos pacientes e menor expressão do fas-L e trail nos leucócitos dos pacientes com TE e MF. Os resultados indicaram associação da expressão do gene PRV-1 com a mutação JAK2V617F e com a expressão dos genes a1, bcl-w, bik, bax, c-iap-2 e trail. Com relação à expressão proteica, BCL-XL estava aumentada e TRAIL diminuída em leucócitos de pacientes com MF enquanto que a molécula BID estava diminuída em leucócitos de pacientes com TE. Os miRNA26a, let 7d e miR15a estavam mais expressos nos leucócitos de pacientes com TE e MF. Em pacientes com TE o miR130b estava elevado e o mIR16 diminuído, enquanto que nos pacientes com MF o miR198 estava aumentado. Os linfócitos dos pacientes com TE e MF apresentaram maior resistência à apoptose estimulada por: actinomicina, etoposídeo, teniposídeo, citarabina e estaurosporina do que os linfócitos dos controles. Em conclusão, os dados obtidos sugerem a ligação da alteração do processo de apoptose celular, com a expressão do gene PRV-1 e status da mutação JAK2V617F. Esses resultados contribuem para o melhor entendimento da fisiopatologia das NMPC visto que associam a fisiopatologia da TE e MF com a resistência das células alteradas à apoptose. Essas informações serão úteis no futuro, possibilitando o desenho de novos alvos terapêuticos e a descrição de novos marcadores de prognóstico. / Chronic Myeloproliferative Neoplasms (cMPN) - Essential Thrombocythemia (ET), Primary Myelofibrosis (PMF) and Polycythemia Vera (PV) - are clonal hematopoietic stem cell malignancies characterized by an accumulation of mature myeloid cells in bone marrow and peripheral blood. It seems that apoptotic machinery deregulation contribute to ET and PMF pathogenesis. Despite the advances in the molecular knowledge, the physiopathology of these diseases remains unknown. This study investigated cellular and molecular mechanisms involved in apoptosis process regulation in bone marrow haematopoietic progenitor CD34+ cells and leukocytes from ET and PMF patients. The specifics aims were: (1) to evaluate death receptors family members and Bcl-2 related genes expression as well apoptosis-related microRNAs by Real Time PCR; (2) apoptosis-related protein expression by Western Blot; (3) access mononuclear cells apoptosis resistance by flow cytometry and (4) to correlate the results with JAK2V617F mutation, PRV-1 gene expression and as well clinical data. In ET CD34+ cells, we found overexpression of a1, mcl-1, bid, bok e noxa and a decrease of bax compared to controls, while in leukocytes a1, bcl-2, bcl-w e bcl-xL, bad e bok expression was increased and bid and bimEL expression was lower than in controls. In PMF, a1, bcl-w bak, bok e noxa were overexpressed and bcl-2 downregulated in CD34+ cells. In PMF leukocytes bcl-2, bcl-w e bcl-xL, bad e bok mRNA levels were increased. c-iap-1 was increased in ET and PMF CD34+ cells and c-iap-2 expression elevated in ET and PMF CD34+ cells and leukocytes. Death receptor related genes showed overexpression of fas, fas-L, faim and dr4 in patients CD34+ cells and downregulation of fas-L and trail in ET and PMF leukocytes. We found differential expression of several genes between patients JAK2V617F positive and negative, as well we found correlation between gene expression and JAK2V617F allele burden, white blood cells and platelets count and splenomegaly. PRV-1 gene was overexpressed in ET and PMF leukocytes and showed correlation with JAK2V617F mutation and a1, bcl-w, bik, bax, c-iap-2 and trail gene expression. Regarding protein expression, BCL-XL was increased and TRAIL decreased in PMF leukocytes and BID was decreased in ET leukocytes. miRNA26a, let 7d e miR15a was overexpressed in ET and PMF leukocytes, while miR130b was increased only in ET and miR198 only in PMF. miR16 was downregulated in ET leukocytes comparing to controls. We also detected a resistance to apoptosis-inducers in ET and PMF lymphocytes and we observed correlation between apoptosis percentage and the expression of many studied genes. In conclusion, the results indicate the participations of Bcl-2 family genes and Death Receptor pathway genes, as well PRV-1 and JAK2V617F mutation in these disorders, which contribute to elucidate cMPN physiopathology and might lead to the discovery of new cMPN therapies and molecular markers.
18

Expressão gênica dos transportadores de membrana ABCB1,ABCG2, SLC22A1 e SLCO1A2 em linhagens celulares tratadas com inibidor comercial da via JAK-STAT / Gene expression of drug transporters ABCB1, ABCG2, SLC22A1 and SLCO1A2 in cell lines treated with commercial inhibitor of JAK-STAT pathway.

Gomes, Guilherme Wataru 24 November 2015 (has links)
INTRODUÇÃO: A desregulação da via de sinalização JAK-STAT é uma característica marcante das neoplasias mieloproliferativas (NMPs), doenças clonais da célula tronco hematopoética, dentre as quais encontra-se a mielofibrose (MF). Diversos inibidores de JAK foram desenvolvidos para o tratamento da MF e encontram-se em diferentes fases de desenvolvimento clínico. Devido ao seu desenvolvimento recente, pouco se sabe a respeito do papel de transportadores de membrana na farmacocinética desses compostos. Essas proteínas realizam o influxo e efluxo celular de substratos endógenos e xenobióticos, e alterações na expressão desses transportadores podem influenciar a resposta a esses fármacos. OBJETIVO: Avaliar o efeito de um inibidor comercial da via JAK-STAT na expressão gênica dos transportadores de membrana ABCB1, ABCG2, SLC22A1 e SLCO1A2 em células HepG2, Caco-2 e HEL92.1.7. MÉTODOS: Linhagens de carcinoma hepatocelular (HepG2), adenocarcinoma colorretal (Caco-2) e eritroleucemia humana homozigotas para JAK2V617F (HEL92.1.7) foram cultivadas e tratadas o inibidor comercial da via JAK-STAT JAK Inhibitor I. Para determinar a concentração ideal para o tratamento com o inibidor, as células foram tratadas com diversas concentrações do inibidor de JAK por 24 horas e foram feitos testes de viabilidade celular e fragmentação do DNA. Com as condições de tratamento padronizadas, foi extraído o RNA total das células e sintetizado o cDNA, para análise das expressões de RNAm dos genes ABCB1, ABCG2, SLC22A1 e SLCO1A2 por PCR em tempo real. Foi também avaliada a expressão dos transportadores de efluxo ABCB1 e ABCG2 por citometria de fluxo, utilizando anticorpos primários direcionados a essas proteínas. RESULTADOS: Nas células HepG2, foi observado um aumento da expressão de RNAm de ABCB1 nas células tratadas com 4,00 µM do inibidor de JAK, quando comparado com o controle (células incubadas apenas com o veículo) (P=0,041). Não foi observada alteração da expressão de RNAm de ABCG2 e SLC22A1 com o tratamento com o inibidor de JAK nessa linhagem (P>0,05); a expressão de RNAm de SLCO1A2 não foi detectada nessa linhagem. Nas células Caco-2, a expressão de ABCB1, ABCG2, SLC22A1 e SLCO1A2 não se alterou com o tratamento com o inibidor de JAK nas concentrações utilizadas (0,25 µM a 1,00 µM) por 24 horas (P>0,05). Para as células HEL92.1.7, não foi observada diferença na expressão de RNAm de ABCB1, ABCG2 e SLC22A1 com o tratamento com 1,00 µM do inibidor de JAK por 24 horas em comparação ao controle (P>0,05); nessa linhagem, a expressão de RNAm de SLCO1A2 não foi detectada. A expressão proteica dos transportadores ABCB1 e ABCG2 não sofreu alteração com o tratamento com o inibidor de JAK nas condições utilizadas nas três linhagens celulares estudadas (P>0,05). CONCLUSÕES: Apenas as células HepG2 apresentaram um aumento da expressão de RNAm do transportador de efluxo ABCB1 em concentrações elevadas do inibidor de JAK, sugerindo que os inibidores de JAK podem modular a expressão do gene desse transportador no fígado. O tratamento com o inibidor da via JAK-STAT não foi associado com alterações na expressão proteica de ABCB1 e ABCG2 em todas as células estudadas. / BACKGROUND: JAK-STAT pathway signaling disregulation is a hallmark of myeloproliferative neoplasms (MPN), hematopoietic stem cell clonal diseases, among which is myelofibrosis (MF). Several JAK inhibitors have been developed for MF treatment and are found in different stages of clinical development. Because the recent development of these compounds, the role of drug transporters in their pharmacokinetics is poorly understood. These proteins perform celular influx and effux of endogenous substrates and xenobiotics, and changes in the expression of these drugs transporters may affect the response to these drugs. AIM: To evaluate the effect of a JAK-STAT pathway commercial inhibitor in gene expression of drug transporters ABCB1, ABCG2, SLC22A1 and SLCO1A2 in HepG2, Caco-2 and HEL92.1.7 cells. METHODS: Hepatocellular carcinoma cell line HepG2, colorectal adenocarcinoma cell line Caco-2 and human erythroleukemia homozygous JAK2V617F cell line HEL92.1.7 were grown and treated with the JAK-STAT pathway inhibitor JAK Inhibitor I. In order to determine the optimal concentration for treatment with the inhibitor, cells were treated with several concentrations of JAK inhibitor by 24 hours, and cell viability and DNA fragmentation tests were performed. Once the treatment conditions were standardized, total RNA were obtained from the cells, and cDNA was synthesized in order to evaluate the mRNA expression of ABCB1, ABCG2, SLC22A1 and SLCO1A2 genes, performed by real time PCR. We also evaluate the expression of drug efflux transporters ABCB1 and ABCG2 by flow cytometry, using primary antibodies directed to these proteins. RESULTS: In HepG2 cells, it was observed an increase in ABCB1 mRNA expression in cells treated with 4,00 µM of JAK inhibitor, when compared with controls (cells exposed only to the vehicle) (P=0.041). There was no change in ABCB2 and SLC22A1 mRNA expression with the treatment with JAK inhibitor in this cell line (P>0.05); SLCO1A2 mRNA was not detected in this cell line. In Caco-2 cells, ABCB1, ABCG2, SLC22A1 and SLCO1A2 mRNA expression did not change with treatment with the JAK inhibitor at the concentrations used (0.25 µM to 1.00 µM) by 24 hours (P>0.05). In HEL92.1.7 cells, it was not observed differences in ABCB1, ABCG2 and SLC22A1 mRNA expression with the treatment with 1 µM of JAK inhibitor by 24 hours when compared with controls (P>0.05); in this cell line, SLCO1A2 mRNA was not detected. Protein expression of ABCB1 and ABCG2 drug transporters has not changed with treatment with the JAK inhibitor under the conditions used in the three cell lines studied. CONCLUSIONS: Only HepG2 cells presented an increase in mRNA expression of drug efflux transporter ABCB1 in presence of high levels of JAK inhibitor, suggesting that JAK inhibitors could modulate this transporter gene expression in liver. Treatment with JAK-STAT pathway inhibitor was not associated with changes in ABCB1 and ABCG2 protein expression in all cell lines studied.
19

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
20

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 0.0416 seconds