Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 512(7512): 78-81, 2014 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-25043017

RESUMEN

Myeloproliferative neoplasms (MPNs) are diseases caused by mutations in the haematopoietic stem cell (HSC) compartment. Most MPN patients have a common acquired mutation of Janus kinase 2 (JAK2) gene in HSCs that renders this kinase constitutively active, leading to uncontrolled cell expansion. The bone marrow microenvironment might contribute to the clinical outcomes of this common event. We previously showed that bone marrow nestin(+) mesenchymal stem cells (MSCs) innervated by sympathetic nerve fibres regulate normal HSCs. Here we demonstrate that abrogation of this regulatory circuit is essential for MPN pathogenesis. Sympathetic nerve fibres, supporting Schwann cells and nestin(+) MSCs are consistently reduced in the bone marrow of MPN patients and mice expressing the human JAK2(V617F) mutation in HSCs. Unexpectedly, MSC reduction is not due to differentiation but is caused by bone marrow neural damage and Schwann cell death triggered by interleukin-1ß produced by mutant HSCs. In turn, in vivo depletion of nestin(+) cells or their production of CXCL12 expanded mutant HSC number and accelerated MPN progression. In contrast, administration of neuroprotective or sympathomimetic drugs prevented mutant HSC expansion. Treatment with ß3-adrenergic agonists that restored the sympathetic regulation of nestin(+) MSCs prevented the loss of these cells and blocked MPN progression by indirectly reducing the number of leukaemic stem cells. Our results demonstrate that mutant-HSC-driven niche damage critically contributes to disease manifestation in MPN and identify niche-forming MSCs and their neural regulation as promising therapeutic targets.


Asunto(s)
Células Madre Hematopoyéticas/patología , Trastornos Mieloproliferativos/patología , Neoplasias/patología , Fibras Nerviosas/patología , Nicho de Células Madre , Sistema Nervioso Simpático/patología , Agonistas de Receptores Adrenérgicos beta 3/farmacología , Agonistas de Receptores Adrenérgicos beta 3/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Progresión de la Enfermedad , Femenino , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Interleucina-1beta/metabolismo , Janus Quinasa 2/genética , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/patología , Ratones , Trastornos Mieloproliferativos/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Fibras Nerviosas/efectos de los fármacos , Nestina/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Receptores Adrenérgicos beta 3/metabolismo , Células de Schwann/efectos de los fármacos , Células de Schwann/patología , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiopatología
2.
Haematologica ; 103(10): 1593-1603, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30076180

RESUMEN

Pathological erythropoiesis with consequent anemia is a leading cause of symptomatic morbidity in internal medicine. The etiologies of anemia are complex and include reactive as well as neoplastic conditions. Clonal expansion of erythroid cells in the bone marrow may result in peripheral erythrocytosis and polycythemia but can also result in anemia when clonal cells are dysplastic and have a maturation arrest that leads to apoptosis and hinders migration, a constellation typically seen in the myelodysplastic syndromes. Rarely, clonal expansion of immature erythroid blasts results in a clinical picture resembling erythroid leukemia. Although several mechanisms underlying normal and abnormal erythropoiesis and the pathogenesis of related disorders have been deciphered in recent years, little is known about specific markers and targets through which prognosis and therapy could be improved in anemic or polycythemic patients. In order to discuss new markers, targets and novel therapeutic approaches in erythroid disorders and the related pathologies, a workshop was organized in Vienna in April 2017. The outcomes of this workshop are summarized in this review, which includes a discussion of new diagnostic and prognostic markers, the updated WHO classification, and an overview of new drugs used to stimulate or to interfere with erythropoiesis in various neoplastic and reactive conditions. The use and usefulness of established and novel erythropoiesis-stimulating agents for various indications, including myelodysplastic syndromes and other neoplasms, are also discussed.


Asunto(s)
Anemia/metabolismo , Células Eritroides/metabolismo , Eritropoyesis , Regulación Neoplásica de la Expresión Génica , Neoplasias/metabolismo , Policitemia/metabolismo , Adulto , Anemia/patología , Células Eritroides/patología , Humanos , Neoplasias/patología , Policitemia/patología
3.
Mol Cancer Res ; 21(8): 768-778, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37255411

RESUMEN

Certain arylsulfonamides (ArSulf) induce an interaction between the E3 ligase substrate adaptor DCAF15 and the critical splicing factor RBM39, ultimately causing its degradation. However, degradation of a splicing factor introduces complex pleiotropic effects that are difficult to untangle, since, aside from direct protein degradation, downstream transcriptional effects also influence the proteome. By overlaying transcriptional data and proteome datasets, we distinguish transcriptional from direct degradation effects, pinpointing those proteins most impacted by splicing changes. Using our workflow, we identify and validate the upregulation of the arginine-and-serine rich protein (RSRP1) and the downregulation of the key kinesin motor proteins KIF20A and KIF20B due to altered splicing in the absence of RBM39. We further show that kinesin downregulation is connected to the multinucleation phenotype observed upon RBM39 depletion by ArSulfs. Our approach should be helpful in the assessment of potential cancer drug candidates which target splicing factors. IMPLICATIONS: Our approach provides a workflow for identifying and studying the most strongly modulated proteins when splicing is altered. The work also uncovers a splicing-based approach toward pharmacologic targeting of mitotic kinesins.


Asunto(s)
Cinesinas , Proteínas de Unión al ARN , Humanos , Cinesinas/genética , Cinesinas/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteoma/metabolismo , Unión Proteica , Factores de Empalme de ARN/metabolismo
4.
Nat Commun ; 14(1): 6185, 2023 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-37794021

RESUMEN

The myeloid transcription factor CEBPA is recurrently biallelically mutated (i.e., double mutated; CEBPADM) in acute myeloid leukemia (AML) with a combination of hypermorphic N-terminal mutations (CEBPANT), promoting expression of the leukemia-associated p30 isoform, and amorphic C-terminal mutations. The most frequently co-mutated genes in CEBPADM AML are GATA2 and TET2, however the molecular mechanisms underlying this co-mutational spectrum are incomplete. By combining transcriptomic and epigenomic analyses of CEBPA-TET2 co-mutated patients with models thereof, we identify GATA2 as a conserved target of the CEBPA-TET2 mutational axis, providing a rationale for the mutational spectra in CEBPADM AML. Elevated CEBPA levels, driven by CEBPANT, mediate recruitment of TET2 to the Gata2 distal hematopoietic enhancer thereby increasing Gata2 expression. Concurrent loss of TET2 in CEBPADM AML induces a competitive advantage by increasing Gata2 promoter methylation, thereby rebalancing GATA2 levels. Of clinical relevance, demethylating treatment of Cebpa-Tet2 co-mutated AML restores Gata2 levels and prolongs disease latency.


Asunto(s)
Dioxigenasas , Leucemia Mieloide Aguda , Humanos , Leucemia Mieloide Aguda/patología , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Mutación , Secuencias Reguladoras de Ácidos Nucleicos , Regiones Promotoras Genéticas/genética , Factor de Transcripción GATA2/genética , Factor de Transcripción GATA2/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dioxigenasas/metabolismo
5.
Cell Physiol Biochem ; 30(4): 1083-96, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23202547

RESUMEN

BACKGROUND: PIM1 is a constitutively active serine-threonine kinase regulating cell survival and proliferation. Increased PIM1 expression has been correlated with cancer metastasis by facilitating migration and anti-adhesion. Endothelial cells play a pivotal role in these processes by contributing a barrier to the blood stream. Here, we investigated whether PIM1 regulates mouse aortic endothelial cell (MAEC) monolayer integrity. METHODS: Pim1-/-MAEC were isolated from Pim1 knockout mice and used in trypsinization-, wound closure assays, electrical cell-substrate sensing, immunostaining, cDNA transfection and as RNA source for microarray analysis. RESULTS: Pim1-/-MAEC displayed decreased migration, slowed cell detachment and increased electrical resistance across the endothelial monolayer. Reintroduction of Pim1- cDNA into Pim1-/-MAEC significantly restored wildtype adhesive characteristics. Pim1-/--MAEC displayed enhanced focal adhesion and adherens junction structures containing vinculin and ß-catenin, respectively. Junctional molecules such as Cadherin 13 and matrix components such as Collagen 6a3 were highly upregulated in Pim1-/- cells. Intriguingly, extracellular matrix deposited by Pim1-/- cells alone was sufficient to induce the hyperadhesive phenotype in wildtype endothelial cells. CONCLUSION: Loss of Pim1 induces a strong adhesive phenotype by enhancing endothelial cell-cell and cell-matrix adhesion by the deposition of a specific extracellular matrix. Targeting PIM1 function therefore might be important to promote endothelial barrier integrity.


Asunto(s)
Células Endoteliales/citología , Células Endoteliales/metabolismo , Proteínas Proto-Oncogénicas c-pim-1/genética , Uniones Adherentes/metabolismo , Animales , Aorta/citología , Adhesión Celular , Movimiento Celular , Células Cultivadas , Adhesiones Focales/metabolismo , Eliminación de Gen , Humanos , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Transfección , Cicatrización de Heridas
6.
Biochem Biophys Res Commun ; 429(1-2): 24-30, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23131564

RESUMEN

The PIM serine/threonine kinases and the mTOR/AKT pathway integrate growth factor signaling and promote cell proliferation and survival. They both share phosphorylation targets and have overlapping functions, which can partially substitute for each other. In cancer cells PIM kinases have been reported to produce resistance to mTOR inhibition by rapamycin. Tumor growth depends highly on blood vessel infiltration into the malignant tissue and therefore on endothelial cell proliferation. We therefore investigated how the PIM1 kinase modulates growth inhibitory effects of rapamycin in mouse aortic endothelial cells (MAEC). We found that proliferation of MAEC lacking Pim1 was significantly more sensitive to rapamycin inhibition, compared to wildtype cells. Inhibition of mTOR and AKT in normal MAEC resulted in significantly elevated PIM1 protein levels in the cytosol and in the nucleus. We observed that truncation of the C-terminal part of Pim1 beyond Ser 276 resulted in almost exclusive nuclear localization of the protein. Re-expression of this Pim1 deletion mutant significantly increased the proliferation of Pim1(-/-) cells when compared to expression of the wildtype Pim1 cDNA. Finally, overexpression of the nuclear localization mutant and the wildtype Pim1 resulted in complete resistance to growth inhibition by rapamycin. Thus, mTOR inhibition-induced nuclear accumulation of PIM1 or expression of a nuclear C-terminal PIM1 truncation mutant is sufficient to increase endothelial cell proliferation, suggesting that nuclear localization of PIM1 is important for resistance of MAEC to rapamycin-mediated inhibition of proliferation.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Núcleo Celular/enzimología , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Sirolimus/farmacología , Animales , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Ratones , Proteínas Proto-Oncogénicas c-pim-1/genética , Eliminación de Secuencia
7.
J Exp Clin Cancer Res ; 41(1): 34, 2022 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-35073946

RESUMEN

BACKGROUND: Acute myeloid leukemia (AML) is characterized by accumulation of aberrantly differentiated hematopoietic myeloid progenitor cells. The karyotyping-silent NUP98-NSD1 fusion is a molecular hallmark of pediatric AML and is associated with the activating FLT3-ITD mutation in > 70% of the cases. NUP98-NSD1 fusion protein promotes myeloid progenitor self-renewal in mice via unknown molecular mechanism requiring both the NUP98 and the NSD1 moieties. METHODS: We used affinity purification coupled to label-free mass spectrometry (AP-MS) to examine the effect of NUP98-NSD1 structural domain deletions on nuclear interactome binding. We determined their functional relevance in NUP98-NSD1 immortalized primary murine hematopoietic stem and progenitor cells (HSPC) by inducible knockdown, pharmacological targeting, methylcellulose assay, RT-qPCR analysis and/or proximity ligation assays (PLA). Fluorescence recovery after photobleaching and b-isoxazole assay were performed to examine the phase transition capacity of NUP98-NSD1 in vitro and in vivo. RESULTS: We show that NUP98-NSD1 core interactome binding is largely dependent on the NUP98 phenylalanine-glycine (FG) repeat domains which mediate formation of liquid-like phase-separated NUP98-NSD1 nuclear condensates. We identified condensate constituents including imitation switch (ISWI) family member SMARCA5 and BPTF (bromodomain PHD finger transcription factor), both members of the nucleosome remodeling factor complex (NURF). We validated the interaction with SMARCA5 in NUP98-NSD1+ patient cells and demonstrated its functional role in NUP98-NSD1/FLT3-ITD immortalized primary murine hematopoietic cells by genetic and pharmacological targeting. Notably, SMARCA5 inhibition did not affect NUP98-NSD1 condensates suggesting that functional activity rather than condensate formation per se is crucial to maintain the transformed phenotype. CONCLUSIONS: NUP98-NSD1 interacts and colocalizes on the genome with SMARCA5 which is an essential mediator of the NUP98-NSD1 transformation in hematopoietic cells. Formation of NUP98-NSD1 phase-separated nuclear condensates is not sufficient for the maintenance of transformed phenotype, which suggests that selective targeting of condensate constituents might represent a new therapeutic strategy for NUP98-NSD1 driven AML.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Hematopoyesis/genética , Proteínas de Complejo Poro Nuclear/metabolismo , Proteómica/métodos , Animales , Humanos , Ratones
8.
Microbiol Spectr ; 10(4): e0147822, 2022 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-35852337

RESUMEN

Moloney murine leukemia virus (MLV) infects BALB/c mice and induces T-cell lymphoma in mice. Retroviral integration is mediated by the interaction of the MLV integrase (IN) with members of the bromodomain and extraterminal motif (BET) protein family (BRD2, BRD3, and BRD4). The introduction of the W390A mutation into MLV IN abolishes the BET interaction. Here, we compared the replication of W390A MLV to that of wild-type (WT) MLV in adult BALB/c mice to study the role of BET proteins in replication, integration, and tumorigenesis in vivo. Comparing WT and W390A MLV infections revealed similar viral loads in the blood, thymus, and spleen cells. Interestingly, W390A MLV integration was retargeted away from GC-enriched genomic regions. However, both WT MLV- and W390A MLV-infected mice developed T-cell lymphoma after similar latencies represented by an enlarged thymus and spleen and multiorgan tumor infiltration. Integration site sequencing from splenic tumor cells revealed clonal expansion in all WT MLV- and W390A MLV-infected mice. However, the integration profiles of W390A MLV and WT MLV differed significantly. Integrations were enriched in enhancers and promoters, but compared to the WT, W390A MLV integrated less frequently into enhancers and more frequently into oncogene bodies such as Notch1 and Ppp1r16b. We conclude that host factors direct MLV in vivo integration site selection. Although BET proteins target WT MLV integration preferentially toward enhancers and promoters, insertional lymphomagenesis can occur independently from BET, likely due to the intrinsically strong enhancer/promoter of the MLV long terminal repeat (LTR). IMPORTANCE In this study, we have shown that the in vivo replication of murine leukemia virus happens independently of BET proteins, which are key host determinants involved in retroviral integration site selection. This finding opens a new research line in the discovery of alternative viral or host factors that may complement the dominant host factor. In addition, our results show that BET-independent murine leukemia virus uncouples insertional mutagenesis from gene enhancers, although lymphomagenesis still occurs despite the lack of an interaction with BET proteins. Our findings also have implications for the engineering of BET-independent MLV-based vectors for gene therapy, which may not be a safe alternative.


Asunto(s)
Linfoma de Células T , Proteínas Nucleares , Animales , Genómica , Integrasas/genética , Integrasas/metabolismo , Virus de la Leucemia Murina/genética , Virus de la Leucemia Murina/metabolismo , Ratones , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Integración Viral/genética
9.
Haematologica ; 95(6): 1004-15, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20145274

RESUMEN

The identification as cooperating targets of Proviral Integrations of Moloney virus in murine lymphomas suggested early on that PIM serine/threonine kinases play an important role in cancer biology. Whereas elevated levels of PIM1 and PIM2 were mostly found in hematologic malignancies and prostate cancer, increased PIM3 expression was observed in different solid tumors. PIM kinases are constitutively active and their activity supports in vitro and in vivo tumor cell growth and survival through modification of an increasing number of common as well as isoform-specific substrates including several cell cycle regulators and apoptosis mediators. PIM1 but not PIM2 seems also to mediate homing and migration of normal and malignant hematopoietic cells by regulating chemokine receptor surface expression. Knockdown experiments by RNA interference or dominant-negative acting mutants suggested that PIM kinases are important for maintenance of a transformed phenotype and therefore potential therapeutic targets. Determination of the protein structure facilitated identification of an increasing number of potent small molecule PIM kinase inhibitors with in vitro and in vivo anticancer activity. Ongoing efforts aim to identify isoform-specific PIM inhibitors that would not only help to dissect the kinase function but hopefully also provide targeted therapeutics. Here, we summarize the current knowledge about the role of PIM serine/threonine kinases for the pathogenesis and therapy of hematologic malignancies and solid cancers, and we highlight structural principles and recent progress on small molecule PIM kinase inhibitors that are on their way into first clinical trials.


Asunto(s)
Neoplasias Hematológicas/genética , Neoplasias Hematológicas/terapia , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-pim-1/química , Proteínas Proto-Oncogénicas c-pim-1/genética , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/genética , Secuencia de Aminoácidos , Animales , Neoplasias Hematológicas/enzimología , Humanos , Datos de Secuencia Molecular , Neoplasias/enzimología , Neoplasias/genética , Neoplasias/terapia , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores
10.
Hemasphere ; 2(3): e43, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31723771

RESUMEN

ZEB1 and ZEB2 play pivotal roles in solid cancer metastasis by allowing cancer cells to invade and disseminate through the transcriptional regulation of epithelial-to-mesenchymal transition. ZEB expression is also associated with the acquisition of cancer stem cell properties and therapy resistance. Consequently, expression levels of ZEB1/2 and of their direct target genes are widely seen as reliable prognostic markers for solid tumor aggressiveness and cancer patient outcome. Recent loss-of-function mouse models demonstrated that both ZEBs are also essential hematopoietic transcription factors governing blood lineage commitment and fidelity. Interestingly, both gain- and loss-of-function mutations have been reported in multiple hematological malignancies. Combined with emerging functional studies, these data suggest that ZEB1 and ZEB2 can act as tumor suppressors and/or oncogenes in blood borne malignancies, depending on the cellular context. Here, we review these novel insights and discuss how balanced expression of ZEB proteins may be essential to safeguard the functionality of the immune system and prevent leukemia.

12.
Cancer Cell ; 31(3): 452-465, 2017 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-28292442

RESUMEN

Chimeric transcription factors are a hallmark of human leukemia, but the molecular mechanisms by which they block differentiation and promote aberrant self-renewal remain unclear. Here, we demonstrate that the ETO2-GLIS2 fusion oncoprotein, which is found in aggressive acute megakaryoblastic leukemia, confers megakaryocytic identity via the GLIS2 moiety while both ETO2 and GLIS2 domains are required to drive increased self-renewal properties. ETO2-GLIS2 directly binds DNA to control transcription of associated genes by upregulation of expression and interaction with the ETS-related ERG protein at enhancer elements. Importantly, specific interference with ETO2-GLIS2 oligomerization reverses the transcriptional activation at enhancers and promotes megakaryocytic differentiation, providing a relevant interface to target in this poor-prognosis pediatric leukemia.


Asunto(s)
Leucemia Megacarioblástica Aguda/patología , Proteínas de Fusión Oncogénica/fisiología , Activación Transcripcional , Animales , Diferenciación Celular , Niño , Elementos de Facilitación Genéticos , Factor de Transcripción GATA1/genética , Humanos , Ratones , Proteínas de Fusión Oncogénica/química , Regulador Transcripcional ERG/fisiología
13.
PLoS One ; 11(3): e0152321, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27031510

RESUMEN

Chromosomal translocations involving the nucleoporin NUP98 have been described in several hematopoietic malignancies, in particular acute myeloid leukemia (AML). In the resulting chimeric proteins, Nup98's N-terminal region is fused to the C-terminal region of about 30 different partners, including homeodomain (HD) transcription factors. While transcriptional targets of distinct Nup98 chimeras related to immortalization are relatively well described, little is known about other potential cellular effects of these fusion proteins. By comparing the sub-nuclear localization of a large number of Nup98 fusions with HD and non-HD partners throughout the cell cycle we found that while all Nup98 chimeras were nuclear during interphase, only Nup98-HD fusion proteins exhibited a characteristic speckled appearance. During mitosis, only Nup98-HD fusions were concentrated on chromosomes. Despite the difference in localization, all tested Nup98 chimera provoked morphological alterations in the nuclear envelope (NE), in particular affecting the nuclear lamina and the lamina-associated polypeptide 2α (LAP2α). Importantly, such aberrations were not only observed in transiently transfected HeLa cells but also in mouse bone marrow cells immortalized by Nup98 fusions and in cells derived from leukemia patients harboring Nup98 fusions. Our findings unravel Nup98 fusion-associated NE alterations that may contribute to leukemogenesis.


Asunto(s)
Membrana Nuclear/genética , Membrana Nuclear/patología , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Fusión Oncogénica/genética , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Ciclo Celular , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/metabolismo , Células HeLa , Proteínas de Homeodominio/análisis , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Proteínas de la Membrana/análisis , Proteínas de la Membrana/metabolismo , Ratones , Mitosis , Membrana Nuclear/metabolismo , Proteínas de Complejo Poro Nuclear/análisis , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas de Fusión Oncogénica/análisis , Proteínas de Fusión Oncogénica/metabolismo , Fenotipo , Translocación Genética
14.
Haematologica ; 90(7): 949-68, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15996933

RESUMEN

Over the last decade, major advances have been made in the elucidation of mechanisms involved in leukemogenesis, and this is particularly true with regard to deregulated protein tyrosine kinase (PTK) activation. This progress had led to the development of small molecules that specifically inhibit the abnormally activated kinase. The first example of such targeted therapy is imatinib-mesylate, an inhibitor of the BCR-ABL fusion gene that is found in more than 90% of patients with Philadelphia positive (Ph+) chronic myeloid leukemia (CML) and in 20-30% of those with Ph+ acute lymphoblastic leukemia (ALL). The excellent clinical results obtained with imatinib in CML have completely changed the therapeutic approach to this disease, and imatinib is now the gold standard for treatment of newly diagnosed CML. This has instigated a tremendous effort to develop targeted PTK therapy based on the presence of over 40 chromosomal translocations that lead to deregulation of 12 different PTK associated with various hematologic malignancies. That deregulated PTK are also involved in the pathogenesis of acute leukemia is underlined by the frequent occurrence of mutations leading to constitutive activation of the FLT3. Experimental as well as clinical evidence supports a model of acute leukemia based on the co-operation of constitutive active PTK with mutations of transcriptional regulators. Here we review the general impact of mutated PTK on the pathogenesis of various hematologic malignancies. We also discuss the development of new targeted therapies and strategies to circumvent the increasing problems related to the emergence of drug resistance by targeting downstream signaling mediators that are essential for transformation by deregulated PTK.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias Hematológicas/genética , Mutación , Proteínas Tirosina Quinasas/genética , Transducción de Señal , Animales , Neoplasias Hematológicas/terapia , Humanos , Modelos Biológicos , Tirosina Quinasa 3 Similar a fms/metabolismo
15.
Cancer Res ; 75(23): 5106-5119, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26552700

RESUMEN

The histone acetyltransferases CBP/p300 are involved in recurrent leukemia-associated chromosomal translocations and are key regulators of cell growth. Therefore, efforts to generate inhibitors of CBP/p300 are of clinical value. We developed a specific and potent acetyl-lysine competitive protein-protein interaction inhibitor, I-CBP112, that targets the CBP/p300 bromodomains. Exposure of human and mouse leukemic cell lines to I-CBP112 resulted in substantially impaired colony formation and induced cellular differentiation without significant cytotoxicity. I-CBP112 significantly reduced the leukemia-initiating potential of MLL-AF9(+) acute myeloid leukemia cells in a dose-dependent manner in vitro and in vivo. Interestingly, I-CBP112 increased the cytotoxic activity of BET bromodomain inhibitor JQ1 as well as doxorubicin. Collectively, we report the development and preclinical evaluation of a novel, potent inhibitor targeting CBP/p300 bromodomains that impairs aberrant self-renewal of leukemic cells. The synergistic effects of I-CBP112 and current standard therapy (doxorubicin) as well as emerging treatment strategies (BET inhibition) provide new opportunities for combinatorial treatment of leukemia and potentially other cancers.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Inhibidores Enzimáticos/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Oxazepinas/farmacología , Factores de Transcripción p300-CBP/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacología , Sinergismo Farmacológico , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/química , Humanos , Leucemia Mieloide Aguda/enzimología , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Oxazepinas/administración & dosificación , Oxazepinas/química , Estructura Terciaria de Proteína , Ensayos Antitumor por Modelo de Xenoinjerto , Factores de Transcripción p300-CBP/química
16.
Am J Clin Pathol ; 122(1): 100-5, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15272537

RESUMEN

Distinction of high-grade esthesioneuroblastomas from other poorly differentiated tumors arising in the nasal cavity is an important diagnostic challenge because it determines patient management and prognosis. The human achaete-scute homologue (hASH1) gene is critical in olfactory neuronal differentiation and is expressed in immature olfactory cells; therefore, it could have potential use as a diagnostic marker The aim of the present study was to determine the value of hASH1 messenger RNA (mRNA) levels in differentiating esthesioneuroblastoma from other poorly differentiated tumors. A real-time polymerase chain reaction assay was developed, permitting the comparative determination of hASH1 mRNA levels in triplicate in a double-blind pilot study including 24 frozen cases of esthesioneuroblastoma and poorly differentiated tumors. All 4 positive cases were esthesioneuroblastomas, and all 19 poorly differentiated tumors were negative. In addition, there was an inverse association between the grade of esthesioneuroblastomas and hASH1 mRNA levels. The hASH1 mRNA level might represent a useful tool for distinguishing esthesioneuroblastoma from poorly differentiated tumors of the sinonasal region.


Asunto(s)
Biomarcadores de Tumor/análisis , Proteínas de Unión al ADN/genética , Estesioneuroblastoma Olfatorio/patología , Cavidad Nasal/patología , Neoplasias Nasales/patología , Factores de Transcripción/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diagnóstico Diferencial , Estesioneuroblastoma Olfatorio/genética , Humanos , Inmunohistoquímica , Neoplasias Nasales/genética , Proyectos Piloto , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Cell Stem Cell ; 15(6): 791-804, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-25479752

RESUMEN

Estrogens are potent regulators of mature hematopoietic cells; however, their effects on primitive and malignant hematopoietic cells remain unclear. Using genetic and pharmacological approaches, we observed differential expression and function of estrogen receptors (ERs) in hematopoietic stem cell (HSC) and progenitor subsets. ERα activation with the selective ER modulator (SERM) tamoxifen induced apoptosis in short-term HSCs and multipotent progenitors. In contrast, tamoxifen induced proliferation of quiescent long-term HSCs, altered the expression of self-renewal genes, and compromised hematopoietic reconstitution after myelotoxic stress, which was reversible. In mice, tamoxifen treatment blocked development of JAK2(V617F)-induced myeloproliferative neoplasm in vivo, induced apoptosis of human JAK2(V617F+) HSPCs in a xenograft model, and sensitized MLL-AF9(+) leukemias to chemotherapy. Apoptosis was selectively observed in mutant cells, and tamoxifen treatment only had a minor impact on steady-state hematopoiesis in disease-free animals. Together, these results uncover specific regulation of hematopoietic progenitors by estrogens and potential antileukemic properties of SERMs.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Janus Quinasa 2/metabolismo , Leucemia/metabolismo , Células Progenitoras Mieloides/efectos de los fármacos , Moduladores Selectivos de los Receptores de Estrógeno/administración & dosificación , Tamoxifeno/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Cultivadas , Receptor alfa de Estrógeno/genética , Hematopoyesis/efectos de los fármacos , Hematopoyesis/genética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/fisiología , Humanos , Janus Quinasa 2/genética , Leucemia/tratamiento farmacológico , Leucemia/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Células Progenitoras Mieloides/fisiología , Proteínas de Fusión Oncogénica/metabolismo , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Oncotarget ; 8(53): 90614-90615, 2017 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-29207581
19.
J Med Chem ; 55(1): 403-13, 2012 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-22136433

RESUMEN

Development of both potent and selective kinase inhibitors is a challenging task in modern drug discovery. The innate promiscuity of kinase inhibitors largely results from ATP-mimetic binding to the kinase hinge region. We present a novel class of substituted 7,8-dichloro-1-oxo-ß-carbolines based on the distinct structural features of the alkaloid bauerine C whose kinase inhibitory activity does not rely on canonical ATP-mimetic hinge interactions. Intriguingly, cocrystal structures revealed an unexpected inverted binding mode and the presence of halogen bonds with kinase backbone residues. The compounds exhibit excellent selectivity over a comprehensive panel of human protein kinases while inhibiting selected kinases such as the oncogenic PIM1 at low nanomolar concentrations. Together, our biochemical and structural data suggest that this scaffold may serve as a valuable template for the design and development of specific inhibitors of various kinases including the PIM family of kinases, CLKs, DAPK3 (ZIPK), BMP2K (BIKE), and others.


Asunto(s)
Antineoplásicos/síntesis química , Carbolinas/síntesis química , Modelos Moleculares , Inhibidores de Proteínas Quinasas/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Carbolinas/química , Carbolinas/farmacología , Línea Celular Tumoral , Cristalografía por Rayos X , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Estructura Molecular , Unión Proteica , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Estereoisomerismo , Relación Estructura-Actividad
20.
Cancer Cell ; 19(2): 161-3, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21316597

RESUMEN

JAK2 gain-of-function mutations have been shown to cause myeloproliferative neoplasms. In this issue of Cancer Cell, Liu et al. (2011) demonstrate that these JAK2 mutants, but not wild-type JAK2, directly phosphorylate PRMT5 and inhibit its arginine methyltransferase activity, establishing a link between mutant JAK2 and histone arginine methylation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA