Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Blood Cancer Discov ; 4(1): 12-33, 2023 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-36322781

RESUMO

T-cell acute lymphoblastic leukemia (T-ALL) is a NOTCH1-driven disease in need of novel therapies. Here, we identify a NOTCH1-SIRT1-KAT7 link as a therapeutic vulnerability in T-ALL, in which the histone deacetylase SIRT1 is overexpressed downstream of a NOTCH1-bound enhancer. SIRT1 loss impaired leukemia generation, whereas SIRT1 overexpression accelerated leukemia and conferred resistance to NOTCH1 inhibition in a deacetylase-dependent manner. Moreover, pharmacologic or genetic inhibition of SIRT1 resulted in significant antileukemic effects. Global acetyl proteomics upon SIRT1 loss uncovered hyperacetylation of KAT7 and BRD1, subunits of a histone acetyltransferase complex targeting H4K12. Metabolic and gene-expression profiling revealed metabolic changes together with a transcriptional signature resembling KAT7 deletion. Consistently, SIRT1 loss resulted in reduced H4K12ac, and overexpression of a nonacetylatable KAT7-mutant partly rescued SIRT1 loss-induced proliferation defects. Overall, our results uncover therapeutic targets in T-ALL and reveal a circular feedback mechanism balancing deacetylase/acetyltransferase activation with potentially broad relevance in cancer. SIGNIFICANCE: We identify a T-ALL axis whereby NOTCH1 activates SIRT1 through an enhancer region, and SIRT1 deacetylates and activates KAT7. Targeting SIRT1 shows antileukemic effects, partly mediated by KAT7 inactivation. Our results reveal T-ALL therapeutic targets and uncover a rheostat mechanism between deacetylase/acetyltransferase activities with potentially broader cancer relevance. This article is highlighted in the In This Issue feature, p. 1.


Assuntos
Leucemia de Células T , Leucemia-Linfoma Linfoblástico de Células T Precursoras , Humanos , Transdução de Sinais , Receptor Notch1/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Sirtuína 1/genética , Sirtuína 1/metabolismo , Sirtuína 1/farmacologia , Acetiltransferases/metabolismo , Acetiltransferases/farmacologia , Acetiltransferases/uso terapêutico , Histona Acetiltransferases/metabolismo , Histona Acetiltransferases/farmacologia , Histona Acetiltransferases/uso terapêutico
2.
Proc Natl Acad Sci U S A ; 119(34): e2207009119, 2022 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-35969760

RESUMO

Classical dendritic cells (cDCs) are essential for immune responses and differentiate from hematopoietic stem cells via intermediate progenitors, such as monocyte-DC progenitors (MDPs) and common DC progenitors (CDPs). Upon infection, cDCs are activated and rapidly express host defense-related genes, such as those encoding cytokines and chemokines. Chromatin structures, including nuclear compartments and topologically associating domains (TADs), have been implicated in gene regulation. However, the extent and dynamics of their reorganization during cDC development and activation remain unknown. In this study, we comprehensively determined higher-order chromatin structures by Hi-C in DC progenitors and cDC subpopulations. During cDC differentiation, chromatin activation was initially induced at the MDP stage. Subsequently, a shift from inactive to active nuclear compartments occurred at the cDC gene loci in CDPs, which was followed by increased intra-TAD interactions and loop formation. Mechanistically, the transcription factor IRF8, indispensable for cDC differentiation, mediated chromatin activation and changes into the active compartments in DC progenitors, thereby possibly leading to cDC-specific gene induction. Using an infection model, we found that the chromatin structures of host defense-related gene loci were preestablished in unstimulated cDCs, indicating that the formation of higher-order chromatin structures prior to infection may contribute to the rapid responses to pathogens. Overall, these results suggest that chromatin structure reorganization is closely related to the establishment of cDC-specific gene expression and immune functions. This study advances the fundamental understanding of chromatin reorganization in cDC differentiation and activation.


Assuntos
Montagem e Desmontagem da Cromatina , Células Dendríticas , Células-Tronco Hematopoéticas , Animais , Diferenciação Celular/genética , Cromatina/genética , Cromatina/metabolismo , Células Dendríticas/citologia , Regulação da Expressão Gênica , Camundongos
3.
Genome Res ; 31(12): 2236-2248, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34799402

RESUMO

Alu are high copy number interspersed repeats that have accumulated near genes during primate and human evolution. They are a pervasive source of structural variation in modern humans. Impacts that Alu insertions may have on gene expression are not well understood, although some have been associated with expression quantitative trait loci (eQTLs). Here, we directly test regulatory effects of polymorphic Alu insertions in isolation of other variants on the same haplotype. To screen insertion variants for those with such effects, we used ectopic luciferase reporter assays and evaluated 110 Alu insertion variants, including more than 40 with a potential role in disease risk. We observed a continuum of effects with significant outliers that up- or down-regulate luciferase activity. Using a series of reporter constructs, which included genomic context surrounding the Alu, we can distinguish between instances in which the Alu disrupts another regulator and those in which the Alu introduces new regulatory sequence. We next focused on three polymorphic Alu loci associated with breast cancer that display significant effects in the reporter assay. We used CRISPR to modify the endogenous sequences, establishing cell lines varying in the Alu genotype. Our findings indicate that Alu genotype can alter expression of genes implicated in cancer risk, including PTHLH, RANBP9, and MYC These data show that commonly occurring polymorphic Alu elements can alter transcript levels and potentially contribute to disease risk.

4.
Blood Cancer Discov ; 2(1): 92-109, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33458694

RESUMO

Long-range oncogenic enhancers play an important role in cancer. Yet, whether similar regulation of tumor suppressor genes is relevant remains unclear. Loss of expression of PTEN is associated with the pathogenesis of various cancers, including T-cell leukemia (T-ALL). Here, we identify a highly conserved distal enhancer (PE) that interacts with the PTEN promoter in multiple hematopoietic populations, including T-cells, and acts as a hub of relevant transcription factors in T-ALL. Consistently, loss of PE leads to reduced PTEN levels in T-ALL cells. Moreover, PE-null mice show reduced Pten levels in thymocytes and accelerated development of NOTCH1-induced T-ALL. Furthermore, secondary loss of PE in established leukemias leads to accelerated progression and a gene expression signature driven by Pten loss. Finally, we uncovered recurrent deletions encompassing PE in T-ALL, which are associated with decreased PTEN levels. Altogether, our results identify PE as the first long-range tumor suppressor enhancer directly implicated in cancer.


Assuntos
Elementos Facilitadores Genéticos , PTEN Fosfo-Hidrolase , Leucemia-Linfoma Linfoblástico de Células T Precursoras , Receptor Notch1 , Animais , Diferenciação Celular , Genes Supressores de Tumor , Camundongos , PTEN Fosfo-Hidrolase/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Receptor Notch1/genética , Transdução de Sinais
5.
Cancer Discov ; 9(12): 1774-1791, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31519704

RESUMO

Long-range enhancers govern the temporal and spatial control of gene expression; however, the mechanisms that regulate enhancer activity during normal and malignant development remain poorly understood. Here, we demonstrate a role for aberrant chromatin accessibility in the regulation of MYC expression in T-cell lymphoblastic leukemia (T-ALL). Central to this process, the NOTCH1-MYC enhancer (N-Me), a long-range T cell-specific MYC enhancer, shows dynamic changes in chromatin accessibility during T-cell specification and maturation and an aberrant high degree of chromatin accessibility in mouse and human T-ALL cells. Mechanistically, we demonstrate that GATA3-driven nucleosome eviction dynamically modulates N-Me enhancer activity and is strictly required for NOTCH1-induced T-ALL initiation and maintenance. These results directly implicate aberrant regulation of chromatin accessibility at oncogenic enhancers as a mechanism of leukemic transformation. SIGNIFICANCE: MYC is a major effector of NOTCH1 oncogenic programs in T-ALL. Here, we show a major role for GATA3-mediated enhancer nucleosome eviction as a driver of MYC expression and leukemic transformation. These results support the role of aberrant chromatin accessibility and consequent oncogenic MYC enhancer activation in NOTCH1-induced T-ALL.This article is highlighted in the In This Issue feature, p. 1631.


Assuntos
Elementos Facilitadores Genéticos , Fator de Transcrição GATA3/metabolismo , Leucemia de Células T/patologia , Nucleossomos/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Animais , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Células Jurkat , Leucemia de Células T/genética , Leucemia de Células T/metabolismo , Camundongos , Transplante de Neoplasias , Receptor Notch1/metabolismo
6.
Cell Rep ; 21(5): 1267-1280, 2017 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-29091765

RESUMO

Low-grade astrocytomas (LGAs) carry neomorphic mutations in isocitrate dehydrogenase (IDH) concurrently with P53 and ATRX loss. To model LGA formation, we introduced R132H IDH1, P53 shRNA, and ATRX shRNA into human neural stem cells (NSCs). These oncogenic hits blocked NSC differentiation, increased invasiveness in vivo, and led to a DNA methylation and transcriptional profile resembling IDH1 mutant human LGAs. The differentiation block was caused by transcriptional silencing of the transcription factor SOX2 secondary to disassociation of its promoter from a putative enhancer. This occurred because of reduced binding of the chromatin organizer CTCF to its DNA motifs and disrupted chromatin looping. Our human model of IDH mutant LGA formation implicates impaired NSC differentiation because of repression of SOX2 as an early driver of gliomagenesis.


Assuntos
Isocitrato Desidrogenase/genética , Fatores de Transcrição SOXB1/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Nuclear Ligada ao X/genética , Animais , Apoptose , Astrocitoma/metabolismo , Astrocitoma/patologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Fator de Ligação a CCCTC/metabolismo , Diferenciação Celular , Células Cultivadas , Metilação de DNA , Epigênese Genética , Humanos , Isocitrato Desidrogenase/metabolismo , Camundongos , Camundongos SCID , Gradação de Tumores , Invasividade Neoplásica , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Interferência de RNA , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo , Proteína Nuclear Ligada ao X/antagonistas & inibidores , Proteína Nuclear Ligada ao X/metabolismo
7.
Cell Rep ; 19(8): 1586-1601, 2017 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-28538178

RESUMO

Immunodeficiency is one of the most important causes of mortality associated with Wolf-Hirschhorn syndrome (WHS), a severe rare disease originated by a deletion in chromosome 4p. The WHS candidate 1 (WHSC1) gene has been proposed as one of the main genes responsible for many of the alterations in WHS, but its mechanism of action is still unknown. Here, we present in vivo genetic evidence showing that Whsc1 plays an important role at several points of hematopoietic development. Particularly, our results demonstrate that both differentiation and function of Whsc1-deficient B cells are impaired at several key developmental stages due to profound molecular defects affecting B cell lineage specification, commitment, fitness, and proliferation, demonstrating a causal role for WHSC1 in the immunodeficiency of WHS patients.


Assuntos
Linfócitos B/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Síndrome de Wolf-Hirschhorn/metabolismo , Animais , Apoptose , Ciclo Celular , Diferenciação Celular , Proliferação de Células , Replicação do DNA , Centro Germinativo/citologia , Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Heterozigoto , Camundongos , Recombinação Genética/genética , Estresse Fisiológico
8.
Cell Stem Cell ; 19(6): 784-799, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27570068

RESUMO

Hematopoietic-specific transcription factors require coactivators to communicate with the general transcription machinery and establish transcriptional programs that maintain hematopoietic stem cell (HSC) self-renewal, promote differentiation, and prevent malignant transformation. Mediator is a large coactivator complex that bridges enhancer-localized transcription factors with promoters, but little is known about Mediator function in adult stem cell self-renewal and differentiation. We show that MED12, a member of the Mediator kinase module, is an essential regulator of HSC homeostasis, as in vivo deletion of Med12 causes rapid bone marrow aplasia leading to acute lethality. Deleting other members of the Mediator kinase module does not affect HSC function, suggesting kinase-independent roles of MED12. MED12 deletion destabilizes P300 binding at lineage-specific enhancers, resulting in H3K27Ac depletion, enhancer de-activation, and consequent loss of HSC stemness signatures. As MED12 mutations have been described recently in blood malignancies, alterations in MED12-dependent enhancer regulation may control both physiological and malignant hematopoiesis.


Assuntos
Elementos Facilitadores Genéticos/genética , Hematopoese , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Complexo Mediador/metabolismo , Animais , Apoptose/genética , Medula Óssea/patologia , Sobrevivência Celular/genética , Cromatina/metabolismo , Deleção de Genes , Perfilação da Expressão Gênica , Camundongos , Ligação Proteica , Fatores de Transcrição/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo
9.
Nucleic Acids Res ; 44(18): 8714-8725, 2016 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-27439714

RESUMO

Use of low resolution single cell DNA FISH and population based high resolution chromosome conformation capture techniques have highlighted the importance of pairwise chromatin interactions in gene regulation. However, it is unlikely that associations involving regulatory elements act in isolation of other interacting partners that also influence their impact. Indeed, the influence of multi-loci interactions remains something of an enigma as beyond low-resolution DNA FISH we do not have the appropriate tools to analyze these. Here we present a method that uses standard 4C-seq data to identify multi-loci interactions from the same cell. We demonstrate the feasibility of our method using 4C-seq data sets that identify known pairwise and novel tri-loci interactions involving the Tcrb and Igk antigen receptor enhancers. We further show that the three Igk enhancers, MiEκ, 3'Eκ and Edκ, interact simultaneously in this super-enhancer cluster, which add to our previous findings showing that loss of one element decreases interactions between all three elements as well as reducing their transcriptional output. These findings underscore the functional importance of simultaneous interactions and provide new insight into the relationship between enhancer elements. Our method opens the door for studying multi-loci interactions and their impact on gene regulation in other biological settings.


Assuntos
Cromossomos/metabolismo , Loci Gênicos , Conformação de Ácido Nucleico , Análise de Sequência de DNA/métodos , Cromatina/metabolismo , Elementos Facilitadores Genéticos , Receptor beta de Estrogênio/metabolismo , Genoma , Receptores de Antígenos de Linfócitos T alfa-beta
10.
Cell Rep ; 16(1): 48-55, 2016 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-27320916

RESUMO

During class switch recombination (CSR), B cells replace the Igh Cµ or δ exons with another downstream constant region exon (CH), altering the antibody isotype. CSR occurs through the introduction of AID-mediated double-strand breaks (DSBs) in switch regions and subsequent ligation of broken ends. Here, we developed an assay to investigate the dynamics of DSB formation in individual cells. We demonstrate that the upstream switch region Sµ is first targeted during recombination and that the mechanism underlying this control relies on 53BP1. Surprisingly, regulation of break order occurs through residual binding of 53BP1 to chromatin before the introduction of damage and independent of its established role in DNA repair. Using chromosome conformation capture, we show that 53BP1 mediates changes in chromatin architecture that affect break order. Finally, our results explain how changes in Igh architecture in the absence of 53BP1 could promote inversional rearrangements that compromise CSR.


Assuntos
Quebras de DNA de Cadeia Dupla , Switching de Imunoglobulina/genética , Cadeias Pesadas de Imunoglobulinas/genética , Recombinação Genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo , Animais , Sequência de Bases , Cromatina/metabolismo , Camundongos , Ligação Proteica , Análise de Célula Única
11.
J Exp Med ; 212(11): 1833-50, 2015 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-26438359

RESUMO

The cohesin complex (consisting of Rad21, Smc1a, Smc3, and Stag2 proteins) is critically important for proper sister chromatid separation during mitosis. Mutations in the cohesin complex were recently identified in a variety of human malignancies including acute myeloid leukemia (AML). To address the potential tumor-suppressive function of cohesin in vivo, we generated a series of shRNA mouse models in which endogenous cohesin can be silenced inducibly. Notably, silencing of cohesin complex members did not have a deleterious effect on cell viability. Furthermore, knockdown of cohesin led to gain of replating capacity of mouse hematopoietic progenitor cells. However, cohesin silencing in vivo rapidly altered stem cells homeostasis and myelopoiesis. Likewise, we found widespread changes in chromatin accessibility and expression of genes involved in myelomonocytic maturation and differentiation. Finally, aged cohesin knockdown mice developed a clinical picture closely resembling myeloproliferative disorders/neoplasms (MPNs), including varying degrees of extramedullary hematopoiesis (myeloid metaplasia) and splenomegaly. Our results represent the first successful demonstration of a tumor suppressor function for the cohesin complex, while also confirming that cohesin mutations occur as an early event in leukemogenesis, facilitating the potential development of a myeloid malignancy.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Proteínas Cromossômicas não Histona/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Homeostase , Transtornos Mieloproliferativos/etiologia , Proteínas Supressoras de Tumor/fisiologia , Adulto , Animais , Proteínas de Ciclo Celular/genética , Diferenciação Celular , Cromatina/fisiologia , Proteínas Cromossômicas não Histona/genética , Humanos , Camundongos , Coesinas
12.
Curr Opin Cell Biol ; 25(3): 365-71, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23478218

RESUMO

Translocations occur through the aberrant joining of large stretches of non-contiguous chromosomal regions. The substrates for these illegitimate rearrangements can arise as a result of damage incurred during normal cellular processes, such as transcription and replication, or through the action of genotoxic agents. In lymphocytes many translocations bear signs of having originated from abnormalities introduced during programmed recombination. Although recombination is tightly controlled at different levels, mistakes can occur leading to cytogenetic anomalies that include deletions, insertions, amplifications and translocations, which are an underlying cause of leukemias and lymphomas. In this review we focus on recent studies that provide insight into the origins of translocations that arise during the two lymphocyte specific programmed recombination events: V(D)J and class switch recombination (CSR).


Assuntos
Linfócitos B/metabolismo , Switching de Imunoglobulina , Linfócitos T/metabolismo , Translocação Genética , Recombinação V(D)J , Animais , Linfócitos B/citologia , Quebras de DNA de Cadeia Dupla , Dano ao DNA , Humanos , Recombinação Genética
13.
Mol Cell ; 47(6): 873-85, 2012 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-22864115

RESUMO

Class switch recombination (CSR) has the potential to generate genomic instability in B cells as activation-induced cytidine deaminase (AID), which mediates this process, is known to target many sites outside Igh. Nonetheless we do not fully understand what factors influence AID targeting genome-wide. Given that errors in CSR can lead to dangerous, oncogenic chromosomal translocations it is important to identify the elements that determine which genes are at risk of being "hit" and could be involved in aberrant rearrangements. Here we have investigated the influence of nuclear organization in determining "off-target" activity and the choice of fusion partners. Our studies indicate that the vast majority of known AID-mediated Igh translocation partners are found in chromosomal domains that contact this locus during class switching. Further, these interaction domains can be used to identify other genes that are hit by AID.


Assuntos
Linfócitos B/citologia , Citidina Desaminase/metabolismo , Genes de Cadeia Pesada de Imunoglobulina , Switching de Imunoglobulina , Translocação Genética , Animais , Linfócitos B/metabolismo , Citidina Desaminase/genética , Instabilidade Genômica , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Hipermutação Somática de Imunoglobulina
14.
Birth Defects Res A Clin Mol Teratol ; 88(8): 626-32, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20589884

RESUMO

BACKGROUND: A precise temporal and spatial regulation of gene expression is necessary to achieve neural tube closure. Med12, a subunit of the mediator complex, can bind transcription factors and modulate expression of their target genes. Med12 is essential during early mouse development and is important for neural tube closure. METHODS: We have made use of a mouse line carrying a conditional null allele of the X-linked Med12 gene to generate heterozygous female embryos that express Med12 in a mosaic fashion thus allowing the study of the role of Med12 during neural tube closure. RESULTS: Mosaic expression of Med12 causes a wide variety of embryonic phenotypes. Some embryos were unable to complete turning and were found with arrested development at embryonic day (ED) 9.5. Others were able to pass ED 12.5 and displayed defects in neural tube closure. These defects included exencephaly, spina bifida, craniorachischisis, split face, and curly tail. Histologic and skeletal analyses of these mutant females show that the neural plate is unable to elevate and is completely flat in the regions of the body axis where neural tube closure fails. CONCLUSIONS: We report examples of all known neural tube defects implying Med12 in the full process of neural tube closure along the complete body axis. Our work points to Med12 being an essential coregulator of transcription factors controlling neural tube closure.


Assuntos
Complexo Mediador/genética , Mosaicismo , Defeitos do Tubo Neural/genética , Disrafismo Espinal/genética , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Complexo Mediador/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Gravidez
15.
Mol Cancer ; 8: 121, 2009 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-20003443

RESUMO

BACKGROUND: Over recent years, enzymes of the aldo-keto reductase (AKR) 1C subfamily have been implicated in the progression of prostate, breast, endometrial and leukemic cancers. This is due to the ability of AKR1C enzymes to modify androgens, estrogens, progesterone and prostaglandins (PGs) in a tissue-specific manner, regulating the activity of nuclear receptors and other downstream effects. Evidence supporting a role for AKR1C enzymes in cancer derives mostly from studies with isolated primary cells from patients or immortalized cell lines. Mice are ideal organisms for in vivo studies, using knock-out or over-expression strains. However, the functional conservation of AKR1C enzymes between human and mice has yet to be described. RESULTS: In this study, we have characterized and compared the four human (AKR1C1,-1C2, -1C3 and -1C4) and the eight murine (AKR1C6, -1C12, -1C13, -1C14, -1C18, -1C19, -1C20 and -1C21) isoforms in their phylogeny, substrate preference and tissue distribution. We have found divergent evolution between human and murine AKR1C enzymes that was reflected by differing substrate preference. Murine enzymes did not perform the 11beta-ketoreduction of prostaglandin (PG) D2, an activity specific to human AKR1C3 and important in promoting leukemic cell survival. Instead, murine AKR1C6 was able to perform the 9-ketoreduction of PGE2, an activity absent amongst human isoforms. Nevertheless, reduction of the key steroids androstenedione, 5alpha-dihydrotestosterone, progesterone and estrone was found in murine isoforms. However, unlike humans, no AKR1C isoforms were detected in murine prostate, testes, uterus and haemopoietic progenitors. CONCLUSIONS: This study exposes significant lack of phylogenetic and functional homology between human and murine AKR1C enzymes. Therefore, we conclude that mice are not suitable to model the role of AKR1C in human cancers and leukemia.


Assuntos
Oxirredutases do Álcool/metabolismo , Modelos Biológicos , Oxirredutases do Álcool/química , Oxirredutases do Álcool/genética , Aldeído Redutase , Aldo-Ceto Redutases , Animais , Humanos , Camundongos , Filogenia , Prostaglandinas/metabolismo , Especificidade da Espécie , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA