Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Leukemia ; 38(6): 1342-1352, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38491305

RESUMEN

Thrombopoietin (Tpo), which binds to its specific receptor, the Mpl protein, is the major cytokine regulator of megakaryopoiesis and circulating platelet number. Tpo binding to Mpl triggers activation of Janus kinase 2 (Jak2) and phosphorylation of the receptor, as well as activation of several intracellular signalling cascades that mediate cellular responses. Three tyrosine (Y) residues in the C-terminal region of the Mpl intracellular domain have been implicated as sites of phosphorylation required for regulation of major Tpo-stimulated signalling pathways: Mpl-Y565, Mpl-Y599 and Mpl-Y604. Here, we have introduced mutations in the mouse germline and report a consistent physiological requirement for Mpl-Y599, mutation of which resulted in thrombocytopenia, deficient megakaryopoiesis, low hematopoietic stem cell (HSC) number and function, and attenuated responses to myelosuppression. We further show that in models of myeloproliferative neoplasms (MPN), where Mpl is required for pathogenesis, thrombocytosis was dependent on intact Mpl-Y599. In contrast, Mpl-Y565 was required for negative regulation of Tpo responses; mutation of this residue resulted in excess megakaryopoiesis at steady-state and in response to myelosuppression, and exacerbated thrombocytosis associated with MPN.


Asunto(s)
Hematopoyesis , Trastornos Mieloproliferativos , Receptores de Trombopoyetina , Trombopoyetina , Tirosina , Animales , Receptores de Trombopoyetina/metabolismo , Receptores de Trombopoyetina/genética , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , Ratones , Trombopoyetina/metabolismo , Tirosina/metabolismo , Tirosina/genética , Fosforilación , Ratones Endogámicos C57BL , Células Madre Hematopoyéticas/metabolismo , Transducción de Señal , Mutación , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Trombopoyesis/genética
2.
Nat Commun ; 13(1): 1658, 2022 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-35351876

RESUMEN

The process of epigenetic silencing, while fundamentally important, is not yet completely understood. Here we report a replenishable female mouse embryonic stem cell (mESC) system, Xmas, that allows rapid assessment of X chromosome inactivation (XCI), the epigenetic silencing mechanism of one of the two X chromosomes that enables dosage compensation in female mammals. Through a targeted genetic screen in differentiating Xmas mESCs, we reveal that the BAF complex is required to create nucleosome-depleted regions at promoters on the inactive X chromosome during the earliest stages of establishment of XCI. Without this action gene silencing fails. Xmas mESCs provide a tractable model for screen-based approaches that enable the discovery of unknown facets of the female-specific process of XCI and epigenetic silencing more broadly.


Asunto(s)
ARN Largo no Codificante , Inactivación del Cromosoma X , Animales , Cromatina/genética , Compensación de Dosificación (Genética) , Epigénesis Genética , Femenino , Ratones , ARN Largo no Codificante/genética , Cromosoma X/genética , Inactivación del Cromosoma X/genética
3.
Development ; 148(13)2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34121118

RESUMEN

Development of a branching tree in the embryonic lung is crucial for the formation of a fully mature functional lung at birth. Sox9+ cells present at the tip of the primary embryonic lung endoderm are multipotent cells responsible for branch formation and elongation. We performed a genetic screen in murine primary cells and identified aurora kinase b (Aurkb) as an essential regulator of Sox9+ cells ex vivo. In vivo conditional knockout studies confirmed that Aurkb was required for lung development but was not necessary for postnatal growth and the repair of the adult lung after injury. Deletion of Aurkb in embryonic Sox9+ cells led to the formation of a stunted lung that retained the expression of Sox2 in the proximal airways, as well as Sox9 in the distal tips. Although we found no change in cell polarity, we showed that loss of Aurkb or chemical inhibition of Aurkb caused Sox9+ cells to arrest at G2/M, likely responsible for the lack of branch bifurcation. This work demonstrates the power of genetic screens in identifying novel regulators of Sox9+ progenitor cells and lung branching morphogenesis.


Asunto(s)
Aurora Quinasa B/genética , Aurora Quinasa B/metabolismo , Células Madre Embrionarias/metabolismo , Endodermo/metabolismo , Pulmón/embriología , Factor de Transcripción SOX9/metabolismo , Animales , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Noqueados , Organogénesis , Factor de Transcripción SOX9/genética
4.
Biochem J ; 478(13): 2555-2569, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34109974

RESUMEN

Structural maintenance of chromosomes flexible hinge domain-containing 1 (SMCHD1) is an epigenetic regulator that mediates gene expression silencing at targeted sites across the genome. Our current understanding of SMCHD1's molecular mechanism, and how substitutions within SMCHD1 lead to the diseases, facioscapulohumeral muscular dystrophy (FSHD) and Bosma arhinia microphthalmia syndrome (BAMS), are only emerging. Recent structural studies of its two component domains - the N-terminal ATPase and C-terminal SMC hinge - suggest that dimerization of each domain plays a central role in SMCHD1 function. Here, using biophysical techniques, we demonstrate that the SMCHD1 ATPase undergoes dimerization in a process that is dependent on both the N-terminal UBL (Ubiquitin-like) domain and ATP binding. We show that neither the dimerization event, nor the presence of a C-terminal extension past the transducer domain, affect SMCHD1's in vitro catalytic activity as the rate of ATP turnover remains comparable to the monomeric protein. We further examined the functional importance of the N-terminal UBL domain in cells, revealing that its targeted deletion disrupts the localization of full-length SMCHD1 to chromatin. These findings implicate UBL-mediated SMCHD1 dimerization as a crucial step for chromatin interaction, and thereby for promoting SMCHD1-mediated gene silencing.


Asunto(s)
Cromatina/metabolismo , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , Multimerización de Proteína , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Sitios de Unión/genética , Cromatina/genética , Proteínas Cromosómicas no Histona/genética , Células HEK293 , Humanos , Immunoblotting , Microscopía Fluorescente , Mutación , Unión Proteica , Dominios Proteicos , Dispersión del Ángulo Pequeño , Especificidad por Sustrato , Ubiquitina/química , Ubiquitina/metabolismo , Difracción de Rayos X
5.
Immunity ; 54(6): 1338-1351.e9, 2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-33862015

RESUMEN

Despite advances in single-cell multi-omics, a single stem or progenitor cell can only be tested once. We developed clonal multi-omics, in which daughters of a clone act as surrogates of the founder, thereby allowing multiple independent assays per clone. With SIS-seq, clonal siblings in parallel "sister" assays are examined either for gene expression by RNA sequencing (RNA-seq) or for fate in culture. We identified, and then validated using CRISPR, genes that controlled fate bias for different dendritic cell (DC) subtypes. This included Bcor as a suppressor of plasmacytoid DC (pDC) and conventional DC type 2 (cDC2) numbers during Flt3 ligand-mediated emergency DC development. We then developed SIS-skew to examine development of wild-type and Bcor-deficient siblings of the same clone in parallel. We found Bcor restricted clonal expansion, especially for cDC2s, and suppressed clonal fate potential, especially for pDCs. Therefore, SIS-seq and SIS-skew can reveal the molecular and cellular mechanisms governing clonal fate.


Asunto(s)
Células Dendríticas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Animales , Diferenciación Celular/genética , Línea Celular , Linaje de la Célula/genética , Femenino , Expresión Génica/genética , Células HEK293 , Humanos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Células Madre/metabolismo
6.
Sci Signal ; 13(636)2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32546545

RESUMEN

Structural maintenance of chromosomes flexible hinge domain containing 1 (SMCHD1) is an epigenetic regulator in which polymorphisms cause the human developmental disorder, Bosma arhinia micropthalmia syndrome, and the degenerative disease, facioscapulohumeral muscular dystrophy. SMCHD1 is considered a noncanonical SMC family member because its hinge domain is C-terminal, because it homodimerizes rather than heterodimerizes, and because SMCHD1 contains a GHKL-type, rather than an ABC-type ATPase domain at its N terminus. The hinge domain has been previously implicated in chromatin association; however, the underlying mechanism involved and the basis for SMCHD1 homodimerization are unclear. Here, we used x-ray crystallography to solve the three-dimensional structure of the Smchd1 hinge domain. Together with structure-guided mutagenesis, we defined structural features of the hinge domain that participated in homodimerization and nucleic acid binding, and we identified a functional hotspot required for chromatin localization in cells. This structure provides a template for interpreting the mechanism by which patient polymorphisms within the SMCHD1 hinge domain could compromise function and lead to facioscapulohumeral muscular dystrophy.


Asunto(s)
Proteínas Cromosómicas no Histona/química , Multimerización de Proteína , Animales , Proteínas Cromosómicas no Histona/genética , Cristalografía por Rayos X , Ratones , Ácidos Nucleicos/química , Ácidos Nucleicos/metabolismo , Dominios Proteicos , Estructura Cuaternaria de Proteína , Hermanos
7.
Nat Commun ; 11(1): 3013, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32541654

RESUMEN

B lymphoid development is initiated by the differentiation of hematopoietic stem cells into lineage committed progenitors, ultimately generating mature B cells. This highly regulated process generates clonal immunological diversity via recombination of immunoglobulin V, D and J gene segments. While several transcription factors that control B cell development and V(D)J recombination have been defined, how these processes are initiated and coordinated into a precise regulatory network remains poorly understood. Here, we show that the transcription factor ETS Related Gene (Erg) is essential for early B lymphoid differentiation. Erg initiates a transcriptional network involving the B cell lineage defining genes, Ebf1 and Pax5, which directly promotes expression of key genes involved in V(D)J recombination and formation of the B cell receptor. Complementation of Erg deficiency with a productively rearranged immunoglobulin gene rescued B lineage development, demonstrating that Erg is an essential and stage-specific regulator of the gene regulatory network controlling B lymphopoiesis.


Asunto(s)
Linfocitos B/metabolismo , Diferenciación Celular/genética , Células Madre Hematopoyéticas/metabolismo , Linfopoyesis/genética , Proteínas Oncogénicas/genética , Transcripción Genética , Regulador Transcripcional ERG/genética , Animales , Linfocitos B/citología , Linaje de la Célula/genética , Células Cultivadas , Redes Reguladoras de Genes/genética , Células Madre Hematopoyéticas/citología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Oncogénicas/metabolismo , Factor de Transcripción PAX5/genética , Factor de Transcripción PAX5/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Regulador Transcripcional ERG/metabolismo , Recombinación V(D)J/genética
8.
Nat Commun ; 11(1): 3150, 2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32561755

RESUMEN

MLKL is the essential effector of necroptosis, a form of programmed lytic cell death. We have isolated a mouse strain with a single missense mutation, MlklD139V, that alters the two-helix 'brace' that connects the killer four-helix bundle and regulatory pseudokinase domains. This confers constitutive, RIPK3 independent killing activity to MLKL. Homozygous mutant mice develop lethal postnatal inflammation of the salivary glands and mediastinum. The normal embryonic development of MlklD139V homozygotes until birth, and the absence of any overt phenotype in heterozygotes provides important in vivo precedent for the capacity of cells to clear activated MLKL. These observations offer an important insight into the potential disease-modulating roles of three common human MLKL polymorphisms that encode amino acid substitutions within or adjacent to the brace region. Compound heterozygosity of these variants is found at up to 12-fold the expected frequency in patients that suffer from a pediatric autoinflammatory disease, chronic recurrent multifocal osteomyelitis (CRMO).


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Sistema Hematopoyético/patología , Necroptosis/genética , Proteínas Quinasas/genética , Animales , Animales Recién Nacidos , Enfermedades Autoinflamatorias Hereditarias , Humanos , Inflamación/genética , Ratones , Mutación Missense , Osteomielitis/genética , Proteínas Quinasas/metabolismo
9.
Nature ; 577(7789): 266-270, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31827282

RESUMEN

Acute myeloid leukaemia (AML) is a heterogeneous disease characterized by transcriptional dysregulation that results in a block in differentiation and increased malignant self-renewal. Various epigenetic therapies aimed at reversing these hallmarks of AML have progressed into clinical trials, but most show only modest efficacy owing to an inability to effectively eradicate leukaemia stem cells (LSCs)1. Here, to specifically identify novel dependencies in LSCs, we screened a bespoke library of small hairpin RNAs that target chromatin regulators in a unique ex vivo mouse model of LSCs. We identify the MYST acetyltransferase HBO1 (also known as KAT7 or MYST2) and several known members of the HBO1 protein complex as critical regulators of LSC maintenance. Using CRISPR domain screening and quantitative mass spectrometry, we identified the histone acetyltransferase domain of HBO1 as being essential in the acetylation of histone H3 at K14. H3 acetylated at K14 (H3K14ac) facilitates the processivity of RNA polymerase II to maintain the high expression of key genes (including Hoxa9 and Hoxa10) that help to sustain the functional properties of LSCs. To leverage this dependency therapeutically, we developed a highly potent small-molecule inhibitor of HBO1 and demonstrate its mode of activity as a competitive analogue of acetyl-CoA. Inhibition of HBO1 phenocopied our genetic data and showed efficacy in a broad range of human cell lines and primary AML cells from patients. These biological, structural and chemical insights into a therapeutic target in AML will enable the clinical translation of these findings.


Asunto(s)
Histona Acetiltransferasas/metabolismo , Leucemia Mieloide Aguda/metabolismo , Células Madre Neoplásicas/metabolismo , Animales , Línea Celular Tumoral , Histona Acetiltransferasas/química , Histona Acetiltransferasas/genética , Humanos , Leucemia Mieloide Aguda/genética , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Estructura Terciaria de Proteína
10.
Cell Rep ; 25(7): 1912-1923.e9, 2018 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-30428357

RESUMEN

We and others have recently reported that the SMC protein Smchd1 is a regulator of chromosome conformation. Smchd1 is critical for the structure of the inactive X chromosome and at autosomal targets such as the Hox genes. However, it is unknown how Smchd1 is recruited to these sites. Here, we report that Smchd1 localizes to the inactive X via the Xist-HnrnpK-PRC1 (polycomb repressive complex 1) pathway. Contrary to previous reports, Smchd1 does not bind Xist or other RNA molecules with any specificity. Rather, the localization of Smchd1 to the inactive X is H2AK119ub dependent. Following perturbation of this interaction, Smchd1 is destabilized, which has consequences for gene silencing genome-wide. Our work adds Smchd1 to the PRC1 silencing pathway for X chromosome inactivation.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo K/metabolismo , Complejo Represivo Polycomb 1/metabolismo , ARN Largo no Codificante/metabolismo , Inactivación del Cromosoma X/genética , Animales , Secuencia de Bases , Diferenciación Celular , Femenino , Genoma , Histonas/metabolismo , Lisina/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Oligonucleótidos/metabolismo , Transporte de Proteínas
11.
J Exp Med ; 214(3): 773-791, 2017 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-28190000

RESUMEN

Genetic alterations disrupting the transcription factor IKZF1 (encoding IKAROS) are associated with poor outcome in B lineage acute lymphoblastic leukemia (B-ALL) and occur in >70% of the high-risk BCR-ABL1+ (Ph+) and Ph-like disease subtypes. To examine IKAROS function in this context, we have developed novel mouse models allowing reversible RNAi-based control of Ikaros expression in established B-ALL in vivo. Notably, leukemias driven by combined BCR-ABL1 expression and Ikaros suppression rapidly regress when endogenous Ikaros is restored, causing sustained disease remission or ablation. Comparison of transcriptional profiles accompanying dynamic Ikaros perturbation in murine B-ALL in vivo with two independent human B-ALL cohorts identified nine evolutionarily conserved IKAROS-repressed genes. Notably, high expression of six of these genes is associated with inferior event-free survival in both patient cohorts. Among them are EMP1, which was recently implicated in B-ALL proliferation and prednisolone resistance, and the novel target CTNND1, encoding P120-catenin. We demonstrate that elevated Ctnnd1 expression contributes to maintenance of murine B-ALL cells with compromised Ikaros function. These results suggest that IKZF1 alterations in B-ALL leads to induction of multiple genes associated with proliferation and treatment resistance, identifying potential new therapeutic targets for high-risk disease.


Asunto(s)
Factor de Transcripción Ikaros/fisiología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Animales , Cateninas/genética , Línea Celular Tumoral , Proteínas de Fusión bcr-abl/análisis , Humanos , Proteínas de la Membrana/genética , Ratones , Proteínas de Neoplasias/genética , Proteínas de Unión al ARN/genética , Receptores de Superficie Celular/genética , Catenina delta
13.
Growth Factors ; 32(1): 18-26, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24438083

RESUMEN

Activation of the cell surface receptor, c-Mpl, by the cytokine, thrombopoietin (TPO), underpins megakaryocyte and platelet production in mammals. In humans, mutations in c-Mpl have been identified as the molecular basis of Congenital Amegakaryocytic Thrombocytopenia (CAMT). Here, we show that CAMT-associated mutations in c-Mpl principally lead to defective receptor presentation on the cell surface. In contrast, one CAMT mutant c-Mpl, F104S, was expressed on the cell surface, but showed defective TPO binding and receptor activation. Using mutational analyses, we examined which residues adjacent to F104 within the membrane-distal cytokine receptor homology module (CRM) of c-Mpl comprise the TPO-binding epitope, revealing residues within the predicted Domain 1 E-F and A-B loops and Domain 2 F'-G' loop as key TPO-binding determinants. These studies underscore the importance of the c-Mpl membrane-distal CRM to TPO-binding and suggest that mutations within this CRM that perturb TPO binding could give rise to CAMT.


Asunto(s)
Mutación/genética , Receptores de Trombopoyetina/genética , Trombocitopenia/genética , Trombopoyetina/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Plaquetas/metabolismo , Células COS , Línea Celular , Proliferación Celular , Chlorocebus aethiops , Síndromes Congénitos de Insuficiencia de la Médula Ósea , Megacariocitos/metabolismo , Ratones , Datos de Secuencia Molecular , Proteínas Oncogénicas/genética , Unión Proteica/genética , Estructura Terciaria de Proteína/genética , Receptores de Prolactina/genética , Alineación de Secuencia , Transducción de Señal/genética
14.
PLoS One ; 8(8): e70536, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23990909

RESUMEN

Suppressor of Cytokine Signaling (SOCS)5 is thought to act as a tumour suppressor through negative regulation of JAK/STAT and epidermal growth factor (EGF) signaling. However, the mechanism/s by which SOCS5 acts on these two distinct pathways is unclear. We show for the first time that SOCS5 can interact directly with JAK via a unique, conserved region in its N-terminus, which we have termed the JAK interaction region (JIR). Co-expression of SOCS5 was able to specifically reduce JAK1 and JAK2 (but not JAK3 or TYK2) autophosphorylation and this function required both the conserved JIR and additional sequences within the long SOCS5 N-terminal region. We further demonstrate that SOCS5 can directly inhibit JAK1 kinase activity, although its mechanism of action appears distinct from that of SOCS1 and SOCS3. In addition, we identify phosphoTyr317 in Shc-1 as a high-affinity substrate for the SOCS5-SH2 domain and suggest that SOCS5 may negatively regulate EGF and growth factor-driven Shc-1 signaling by binding to this site. These findings suggest that different domains in SOCS5 contribute to two distinct mechanisms for regulation of cytokine and growth factor signaling.


Asunto(s)
Janus Quinasa 1/metabolismo , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Animales , Sitios de Unión , Citocinas/metabolismo , Vectores Genéticos , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Janus Quinasa 2/metabolismo , Janus Quinasa 3/metabolismo , Ratones , Fosfopéptidos/química , Fosforilación , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Transducción de Señal , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Especificidad por Sustrato , Resonancia por Plasmón de Superficie , TYK2 Quinasa/metabolismo , Dominios Homologos src
15.
J Immunol ; 187(7): 3798-805, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21876038

RESUMEN

The mammalian innate immune system has evolved to recognize foreign molecules derived from pathogens via the TLRs. TLR3 and TLR4 can signal via the TIR domain-containing adapter inducing IFN-ß (TRIF), which results in the transcription of a small array of genes, including IFN-ß. Inducible NO synthase (iNOS), which catalyzes the production of NO, is induced by a range of stimuli, including cytokines and microbes. NO is a potent source of reactive nitrogen species that play an important role in killing intracellular pathogens and forms a crucial component of host defense. We have recently identified iNOS as a target of the mammalian SPSB2 protein. The SOCS box is a peptide motif, which, in conjunction with elongins B and C, recruits cullin-5 and Rbx-2 to form an active E3 ubiquitin ligase complex. In this study, we show that SPSB1 is the only SPSB family member to be regulated by the same TLR pathways that induce iNOS expression and characterize the interaction between SPSB1 and iNOS. Through the use of SPSB1 transgenic mouse macrophages and short hairpin RNA knockdown of SPSB1, we show that SPSB1 controls both the induction of iNOS and the subsequent production of NO downstream of TLR3 and TLR4. Further, we demonstrate that regulation of iNOS by SPSB1 is dependent on the proteasome. These results suggest that SPSB1 acts through a negative-feedback loop that, together with SPSB2, controls the extent of iNOS induction and NO production.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Macrófagos/metabolismo , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Transducción de Señal/inmunología , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Receptores Toll-Like/metabolismo , Animales , Western Blotting , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Proteínas de Unión al ADN/metabolismo , Expresión Génica , Inmunoprecipitación , Macrófagos/inmunología , Ratones , Ratones Transgénicos , Óxido Nítrico/biosíntesis , Óxido Nítrico/inmunología , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/inmunología , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/inmunología , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología
16.
Nucleic Acids Res ; 39(17): 7415-27, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21652639

RESUMEN

Transcriptional control is dependent on a vast network of epigenetic modifications. One epigenetic mark of particular interest is tri-methylation of lysine 27 on histone H3 (H3K27me3), which is catalysed and maintained by Polycomb Repressive Complex 2 (PRC2). Although this histone mark is studied widely, the precise relationship between its local pattern of enrichment and regulation of gene expression is currently unclear. We have used ChIP-seq to generate genome-wide maps of H3K27me3 enrichment, and have identified three enrichment profiles with distinct regulatory consequences. First, a broad domain of H3K27me3 enrichment across the body of genes corresponds to the canonical view of H3K27me3 as inhibitory to transcription. Second, a peak of enrichment around the transcription start site (TSS) is commonly associated with 'bivalent' genes, where H3K4me3 also marks the TSS. Finally and most surprisingly, we identified an enrichment profile with a peak in the promoter of genes that is associated with active transcription. Genes with each of these three profiles were found in different proportions in each of the cell types studied. The data analysis techniques developed here will be useful for the identification of common enrichment profiles for other histone modifications that have important consequences for transcriptional regulation.


Asunto(s)
Histonas/metabolismo , Transcripción Genética , Animales , Células Cultivadas , Inmunoprecipitación de Cromatina , Análisis por Conglomerados , Histonas/química , Lisina/metabolismo , Metilación , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Análisis de Secuencia de ADN , Sitio de Iniciación de la Transcripción
17.
Genes Dev ; 25(3): 251-62, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21245161

RESUMEN

Although many genes are known to be critical for early hematopoiesis in the embryo, it remains unclear whether distinct regulatory pathways exist to control hematopoietic specification versus hematopoietic stem cell (HSC) emergence and function. Due to their interaction with key regulators of hematopoietic commitment, particular interest has focused on the role of the ETS family of transcription factors; of these, ERG is predicted to play an important role in the initiation of hematopoiesis, yet we do not know if or when ERG is required. Using in vitro and in vivo models of hematopoiesis and HSC development, we provide strong evidence that ERG is at the center of a distinct regulatory program that is not required for hematopoietic specification or differentiation but is critical for HSC maintenance during embryonic development. We show that, from the fetal period, ERG acts as a direct upstream regulator of Gata2 and Runx1 gene activity. Without ERG, physiological HSC maintenance fails, leading to the rapid exhaustion of definitive hematopoiesis.


Asunto(s)
Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Proteínas Oncogénicas/metabolismo , Animales , Células Cultivadas , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Factor de Transcripción GATA2/metabolismo , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Oncogénicas/genética , Factores de Transcripción , Regulador Transcripcional ERG
18.
Proc Natl Acad Sci U S A ; 107(50): 21689-94, 2010 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-21115812

RESUMEN

Thrombopoietin (TPO), acting through its receptor Mpl, has two major physiological roles: ensuring production of sufficient platelets via stimulation of megakaryocyte production and maintaining hematopoietic stem cell (HSC) quiescence. Mpl also controls circulating TPO concentration via receptor-mediated internalization and degradation. Here, we demonstrate that the megakaryocytosis and increased platelet mass in mice with mutations in the Myb or p300 genes causes reduced circulating TPO concentration and TPO starvation of the stem-cell compartment, which is exacerbated because these cells additionally exhibit impaired responsiveness to TPO. HSCs from Myb(Plt4/Plt4) mice show altered expression of TPO-responsive genes and, like HSCs from Tpo and Mpl mutant mice, exhibit increased cycling and a decline in the number of HSCs with age. These studies suggest that disorders of platelet number can have profound effects on the HSC compartment via effects on the feedback regulation of circulating TPO concentration.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Animales , Plaquetas/metabolismo , Diferenciación Celular/fisiología , Proteína p300 Asociada a E1A/genética , Proteína p300 Asociada a E1A/metabolismo , Células Madre Hematopoyéticas/citología , Megacariocitos/citología , Megacariocitos/fisiología , Ratones , Ratones Noqueados , Análisis por Micromatrices , Proteínas Proto-Oncogénicas c-myb/genética , Proteínas Proto-Oncogénicas c-myb/metabolismo , Receptores de Trombopoyetina/metabolismo , Trombopoyetina/sangre
19.
Proc Natl Acad Sci U S A ; 107(38): 16625-30, 2010 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-20823251

RESUMEN

With the notable exception of humans, uric acid is degraded to (S)-allantoin in a biochemical pathway catalyzed by urate oxidase, 5-hydroxyisourate (HIU) hydrolase, and 2-oxo-4-hydroxy-4-carboxy-5-ureidoimidazoline decarboxylase in most vertebrate species. A point mutation in the gene encoding mouse HIU hydrolase, Urah, that perturbed uric acid metabolism within the liver was discovered during a mutagenesis screen in mice. The predicted substitution of cysteine for tyrosine in a conserved helical region of the mutant-encoded HIU hydrolase resulted in undetectable protein expression. Mice homozygous for this mutation developed elevated platelet counts secondary to excess thrombopoietin production and hepatomegaly. The majority of homozygous mutant mice also developed hepatocellular carcinoma, and tumor development was accelerated by exposure to radiation. The development of hepatomegaly and liver tumors in mice lacking Urah suggests that uric acid metabolites may be toxic and that urate oxidase activity without HIU hydrolase function may affect liver growth and transformation. The absence of HIU hydrolase in humans predicts slowed metabolism of HIU after clinical administration of exogenous urate oxidase in conditions of uric acid-related pathology. The data suggest that prolonged urate oxidase therapy should be combined with careful assessment of toxicity associated with extrahepatic production of uric acid metabolites.


Asunto(s)
Amidohidrolasas/deficiencia , Amidohidrolasas/genética , Hepatomegalia/enzimología , Hepatomegalia/genética , Neoplasias Hepáticas Experimentales/enzimología , Neoplasias Hepáticas Experimentales/genética , Mutación Puntual , Amidohidrolasas/química , Amidohidrolasas/metabolismo , Secuencia de Aminoácidos , Animales , Femenino , Genes Supresores de Tumor , Hepatocitos/enzimología , Hepatomegalia/etiología , Neoplasias Hepáticas Experimentales/etiología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Trombocitosis/enzimología , Trombocitosis/genética , Trombopoyetina/biosíntesis , Urato Oxidasa/metabolismo , Ácido Úrico/metabolismo , Ácido Úrico/toxicidad
20.
J Cell Biol ; 190(1): 129-41, 2010 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-20603330

RESUMEN

Inducible nitric oxide (NO) synthase (iNOS; NOS2) produces NO and related reactive nitrogen species, which are critical effectors of the innate host response and are required for the intracellular killing of pathogens such as Mycobacterium tuberculosis and Leishmania major. We have identified SPRY domain-containing SOCS (suppressor of cytokine signaling) box protein 2 (SPSB2) as a novel negative regulator that recruits an E3 ubiquitin ligase complex to polyubiquitinate iNOS, resulting in its proteasomal degradation. SPSB2 interacts with the N-terminal region of iNOS via a binding interface on SPSB2 that has been mapped by nuclear magnetic resonance spectroscopy and mutational analyses. SPSB2-deficient macrophages showed prolonged iNOS expression, resulting in a corresponding increase in NO production and enhanced killing of L. major parasites. These results lay the foundation for the development of small molecule inhibitors that could disrupt the SPSB-iNOS interaction and thus prolong the intracellular lifetime of iNOS, which may be beneficial in chronic and persistent infections.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Leishmania major , Leishmaniasis Cutánea/metabolismo , Macrófagos/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Animales , Línea Celular , Proteínas de Unión al ADN/genética , Regulación Enzimológica de la Expresión Génica/genética , Leishmaniasis Cutánea/tratamiento farmacológico , Leishmaniasis Cutánea/genética , Macrófagos/parasitología , Ratones , Ratones Noqueados , Mycobacterium tuberculosis , Óxido Nítrico Sintasa de Tipo II/genética , Complejo de la Endopetidasa Proteasomal/genética , Estructura Terciaria de Proteína , Proteínas Supresoras de la Señalización de Citocinas/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA