Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 138(17): 1540-1553, 2021 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-34086867

RESUMEN

Thalassemia or sickle cell patients with hereditary persistence of fetal hemoglobin (HbF) have an ameliorated clinical phenotype and, in some cases, can achieve transfusion independence. Inactivation via genome editing of γ-globin developmental suppressors, such as BCL11A or LRF/ZBTB7A, or of their binding sites, have been shown to significantly increase expression of endogenous HbF. To broaden the therapeutic window beyond a single-editing approach, we have explored combinations of cis- and trans-editing targets to enhance HbF reactivation. Multiplex mutagenesis in adult CD34+ cells was well tolerated and did not lead to any detectable defect in the cells' proliferation and differentiation, either in vitro or in vivo. The combination of 1 trans and 1 cis mutation resulted in high editing retention in vivo, coupled with almost pancellular HbF expression in NBSGW mice. The greater in vivo performance of this combination was also recapitulated using a novel helper-dependent adenoviral-CRISPR vector (HD-Ad-dualCRISPR) in CD34+ cells from ß-thalassemia patients transplanted to NBSGW mice. A pronounced increase in HbF expression was observed in human red blood cells in mice with established predominant ß0/ß0-thalassemic hemopoiesis after in vivo injection of the HD-Ad-dualCRISPR vector. Collectively, our data suggest that the combination of cis and trans fetal globin reactivation mutations has the potential to significantly increase HbF both totally and on a per cell basis over single editing and could thus provide significant clinical benefit to patients with severe ß-globin phenotype.


Asunto(s)
Antígenos CD34/genética , Hemoglobina Fetal/genética , Mutagénesis , Talasemia beta/genética , Adulto , Animales , Sistemas CRISPR-Cas , Células Cultivadas , Edición Génica , Terapia Genética , Humanos , Ratones , Talasemia beta/terapia , gamma-Globinas/genética
2.
Haematologica ; 108(4): 1053-1067, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35861015

RESUMEN

Although human cell cultures stimulated with dexamethasone suggest that the glucocorticoid receptor (GR) activates stress erythropoiesis, the effects of GR activation on erythropoiesis in vivo remain poorly understood. We characterized the phenotype of a large cohort of patients with Cushing disease, a rare condition associated with elevated cortisol levels. Results from hypercortisolemic patients with active Cushing disease were compared with those obtained from eucortisolemic patients after remission and from volunteers without the disease. Patients with active Cushing disease exhibited erythrocytosis associated with normal hemoglobin F levels. In addition, their blood contained elevated numbers of GR-induced CD163+ monocytes and a unique class of CD34+ cells expressing CD110, CD36, CD133 and the GR-target gene CXCR4. When cultured, these CD34+ cells generated similarly large numbers of immature erythroid cells in the presence and absence of dexamethasone, with raised expression of the GR-target gene GILZ. Of interest, blood from patients with Cushing disease in remission maintained high numbers of CD163+ monocytes and, although their CD34+ cells had a normal phenotype, these cells were unresponsive to added dexamethasone. Collectively, these results indicate that chronic exposure to excess glucocorticoids in vivo leads to erythrocytosis by generating erythroid progenitor cells with a constitutively active GR. Although remission rescues the erythrocytosis and the phenotype of the circulating CD34+ cells, a memory of other prior changes is maintained in remission.


Asunto(s)
Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT) , Policitemia , Humanos , Policitemia/etiología , Células Madre Hematopoyéticas/metabolismo , Glucocorticoides/farmacología , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Dexametasona/farmacología , Células Cultivadas
3.
Trends Genet ; 34(12): 927-940, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30287096

RESUMEN

Disorders in hemoglobin (hemoglobinopathies) were the first monogenic diseases to be characterized and remain among the most common and best understood genetic conditions. Moreover, the study of the ß-globin locus provides a textbook example of developmental gene regulation. The fetal γ-globin genes (HBG1/HBG2) are ordinarily silenced around birth, whereupon their expression is replaced by the adult ß-globin genes (HBB primarily and HBD). Over 50 years ago it was recognized that mutations that cause lifelong persistence of fetal γ-globin expression ameliorate the debilitating effects of mutations in ß-globin. Since then, research has focused on therapeutically reactivating the fetal γ-globin genes. Here, we summarize recent discoveries, focusing on the influence of genome editing technologies, including CRISPR-Cas9, and emerging gene therapy approaches.


Asunto(s)
Terapia Genética/tendencias , Hemoglobinopatías/genética , Globinas beta/genética , gamma-Globinas/genética , Adulto , Sistemas CRISPR-Cas/genética , Edición Génica/tendencias , Hemoglobinopatías/sangre , Hemoglobinopatías/patología , Humanos , Mutación , Globinas beta/uso terapéutico , gamma-Globinas/uso terapéutico
4.
Bioessays ; 41(8): e1900041, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31245868

RESUMEN

Transcriptional silencing may not necessarily depend on the continuous residence of a sequence-specific repressor at a control element and may act via a "hit and run" mechanism. Due to limitations in assays that detect transcription factor (TF) binding, such as chromatin immunoprecipitation followed by high-throughput sequencing (ChIP-seq), this phenomenon may be challenging to detect and therefore its prevalence may be underappreciated. To explore this possibility, erythroid gene promoters that are regulated directly by GATA1 in an inducible system are analyzed. It is found that many regulated genes are bound immediately after induction of GATA1 but the residency of GATA1 decreases over time, particularly at repressed genes. Furthermore, it is shown that the repressive mark H3K27me3 is seldom associated with bound repressors, whereas, in contrast, the active (H3K4me3) histone mark is overwhelmingly associated with TF binding. It is hypothesized that during cellular differentiation and development, certain genes are silenced by repressive TFs that subsequently vacate the region. Catching such repressor TFs in the act of silencing via assays such as ChIP-seq is thus a temporally challenging prospect. The use of inducible systems, epitope tags, and alternative techniques may provide opportunities for detecting elusive "hit and run" transcriptional silencing. Also see the video abstract here https://youtu.be/vgrsoP_HF3g.


Asunto(s)
Silenciador del Gen , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Sitios de Unión , Secuenciación de Inmunoprecipitación de Cromatina , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Sitios Genéticos , Histonas/metabolismo , Humanos , Regiones Promotoras Genéticas , Unión Proteica , Activación Transcripcional
5.
Nucleic Acids Res ; 44(7): 3118-30, 2016 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-26673701

RESUMEN

Transcription factors are often regarded as having two separable components: a DNA-binding domain (DBD) and a functional domain (FD), with the DBD thought to determine target gene recognition. While this holds true for DNA bindingin vitro, it appears thatin vivoFDs can also influence genomic targeting. We fused the FD from the well-characterized transcription factor Krüppel-like Factor 3 (KLF3) to an artificial zinc finger (AZF) protein originally designed to target the Vascular Endothelial Growth Factor-A (VEGF-A) gene promoter. We compared genome-wide occupancy of the KLF3FD-AZF fusion to that observed with AZF. AZF bound to theVEGF-Apromoter as predicted, but was also found to occupy approximately 25,000 other sites, a large number of which contained the expected AZF recognition sequence, GCTGGGGGC. Interestingly, addition of the KLF3 FD re-distributes the fusion protein to new sites, with total DNA occupancy detected at around 50,000 sites. A portion of these sites correspond to known KLF3-bound regions, while others contained sequences similar but not identical to the expected AZF recognition sequence. These results show that FDs can influence and may be useful in directing AZF DNA-binding proteins to specific targets and provide insights into how natural transcription factors operate.


Asunto(s)
Proteínas de Unión al ADN/química , Factores de Transcripción/química , Dedos de Zinc , Sitios de Unión , Cromatina/metabolismo , ADN/química , ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Genoma Humano , Células HEK293 , Humanos , Factores de Transcripción de Tipo Kruppel/química , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética
6.
J Biol Chem ; 291(31): 16048-58, 2016 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-27226561

RESUMEN

The Lgals3 gene encodes a multifunctional ß-galactoside-binding protein, galectin-3. Galectin-3 has been implicated in a broad range of biological processes from chemotaxis and inflammation to fibrosis and apoptosis. The role of galectin-3 as a modulator of inflammation has been studied intensively, and recent evidence suggests that it may serve as a protective factor in obesity and other metabolic disorders. Despite considerable interest in galectin-3, little is known about its physiological regulation at the transcriptional level. Here, using knockout mice, chromatin immunoprecipitations, and cellular and molecular analyses, we show that the zinc finger transcription factor Krüppel-like factor 3 (KLF3) directly represses galectin-3 transcription. We find that galectin-3 is broadly up-regulated in KLF3-deficient mouse tissues, that KLF3 occupies regulatory regions of the Lgals3 gene, and that KLF3 directly binds its cognate elements (CACCC boxes) in the galectin-3 promoter and represses its activation in cellular assays. We also provide mechanistic insights into the regulation of Lgals3, demonstrating that C-terminal binding protein (CtBP) is required to drive optimal KLF3-mediated silencing. These findings help to enhance our understanding of how expression of the inflammatory modulator galectin-3 is controlled, opening up avenues for potential therapeutic interventions in the future.


Asunto(s)
Galectina 3/biosíntesis , Silenciador del Gen , Mediadores de Inflamación/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Represoras/metabolismo , Elementos de Respuesta , Transcripción Genética , Animales , Galectina 3/genética , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Ratones Noqueados , Proteínas Represoras/genética
7.
Trends Genet ; 30(1): 18-23, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24138812

RESUMEN

Hemophilia B is a classic, monogenic blood clotting disease caused by mutations in the coagulation factor IX (F9) locus. Although interpreting mutations within the gene itself has been relatively straightforward, ascribing molecular mechanisms to the complete suite of mutations within the promoter region has proven somewhat difficult and has only recently been achieved. These mutations, which are clustered at discrete transcription factor binding sites, dynamically alter the developmental expression of F9 in different ways. They illustrate how single-nucleotide mutations in cis-regulatory regions can have drastic ramifications for the control of gene expression and in some instances be causative of disease. Here we present the human F9 promoter as a model example for which saturation mutation mapping has revealed the mechanisms of its regulation. Moreover, we suggest that the growing number of genome-wide studies of transcription factor activity will accelerate both the discovery and understanding of regulatory polymorphisms and mutations.


Asunto(s)
Factor IX/genética , Hemofilia B/genética , Mutación , Regiones Promotoras Genéticas , Secuencia de Aminoácidos , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Factor IX/metabolismo , Regulación de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Blood ; 126(1): 89-93, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26019277

RESUMEN

Elevated fetal hemoglobin (HbF) ameliorates the clinical severity of hemoglobinopathies such as ß-thalassemia and sickle cell anemia. Currently, the only curative approach for individuals under chronic transfusion/chelation support therapy is allogeneic stem cell transplantation. However, recent analyses of heritable variations in HbF levels have provided a new therapeutic target for HbF reactivation: the transcriptional repressor BCL11A. Erythroid-specific BCL11A abrogation is now actively being sought as a therapeutic avenue, but the specific impact of such disruption in humans remains to be determined. Although single nucleotide polymorphisms in BCL11A erythroid regulatory elements have been reported, coding mutations are scarcer. It is thus of great interest that patients have recently been described with microdeletions encompassing BCL11A. These patients display neurodevelopmental abnormalities, but whether they show increased HbF has not been reported. We have examined the hematological phenotype, HbF levels, and erythroid BCL11A expression in 3 such patients. Haploinsufficiency of BCL11A induces only partial developmental γ-globin silencing. Of greater interest is that a patient with a downstream deletion exhibits reduced BCL11A expression and increased HbF. Novel erythroid-specific regulatory elements in this region may be required for normal erythroid BCL11A expression, whereas loss of separate elements in the developing brain may explain the neurological phenotype.


Asunto(s)
Proteínas Portadoras/genética , Deleción Cromosómica , Cromosomas Humanos Par 2 , Hemoglobina Fetal/metabolismo , Enfermedades del Sistema Nervioso/genética , Proteínas Nucleares/genética , Adolescente , Niño , Femenino , Humanos , Masculino , Enfermedades del Sistema Nervioso/sangre , Proteínas Represoras , Regulación hacia Arriba
9.
J Biol Chem ; 290(13): 8591-605, 2015 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-25659434

RESUMEN

Krüppel-like factor 3 (KLF3/BKLF), a member of the Krüppel-like factor (KLF) family of transcription factors, is a widely expressed transcriptional repressor with diverse biological roles. Although there is considerable understanding of the molecular mechanisms that allow KLF3 to silence the activity of its target genes, less is known about the signal transduction pathways and post-translational modifications that modulate KLF3 activity in response to physiological stimuli. We observed that KLF3 is modified in a range of different tissues and found that the serine/threonine kinase homeodomain-interacting protein kinase 2 (HIPK2) can both bind and phosphorylate KLF3. Mass spectrometry identified serine 249 as the primary phosphorylation site. Mutation of this site reduces the ability of KLF3 to bind DNA and repress transcription. Furthermore, we also determined that HIPK2 can phosphorylate the KLF3 co-repressor C-terminal binding protein 2 (CtBP2) at serine 428. Finally, we found that phosphorylation of KLF3 and CtBP2 by HIPK2 strengthens the interaction between these two factors and increases transcriptional repression by KLF3. Taken together, our results indicate that HIPK2 potentiates the activity of KLF3.


Asunto(s)
Proteínas Portadoras/fisiología , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Oxidorreductasas de Alcohol , Secuencia de Aminoácidos , Animales , Células COS , Chlorocebus aethiops , Proteínas Co-Represoras , ADN/química , Ensayo de Cambio de Movilidad Electroforética , Factores de Transcripción de Tipo Kruppel/química , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH , Fosforilación , Unión Proteica , Procesamiento Proteico-Postraduccional , Transcripción Genética , Activación Transcripcional
10.
Am J Hum Genet ; 92(3): 460-7, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23472758

RESUMEN

Hemophilia B, or the "royal disease," arises from mutations in coagulation factor IX (F9). Mutations within the F9 promoter are associated with a remarkable hemophilia B subtype, termed hemophilia B Leyden, in which symptoms ameliorate after puberty. Mutations at the -5/-6 site (nucleotides -5 and -6 relative to the transcription start site, designated +1) account for the majority of Leyden cases and have been postulated to disrupt the binding of a transcriptional activator, the identity of which has remained elusive for more than 20 years. Here, we show that ONECUT transcription factors (ONECUT1 and ONECUT2) bind to the -5/-6 site. The various hemophilia B Leyden mutations that have been reported in this site inhibit ONECUT binding to varying degrees, which correlate well with their associated clinical severities. In addition, expression of F9 is crucially dependent on ONECUT factors in vivo, and as such, mice deficient in ONECUT1, ONECUT2, or both exhibit depleted levels of F9. Taken together, our findings establish ONECUT transcription factors as the missing hemophilia B Leyden regulators that operate through the -5/-6 site.


Asunto(s)
Factor IX/genética , Hemofilia B/genética , Factor Nuclear 6 del Hepatocito/metabolismo , Proteínas de Homeodominio/metabolismo , Mutación , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Predisposición Genética a la Enfermedad , Células Hep G2 , Humanos , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Transcripción Genética
11.
Nucleic Acids Res ; 42(1): 276-89, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24106088

RESUMEN

Transcription factors (TFs) are often regarded as being composed of a DNA-binding domain (DBD) and a functional domain. The two domains are considered separable and autonomous, with the DBD directing the factor to its target genes and the functional domain imparting transcriptional regulation. We examined an archetypal zinc finger (ZF) TF, Krüppel-like factor 3 with an N-terminal domain that binds the corepressor CtBP and a DBD composed of three ZFs at its C-terminus. We established a system to compare the genomic occupancy profile of wild-type Krüppel-like factor 3 with two mutants affecting the N-terminal functional domain: a mutant unable to contact the cofactor CtBP and a mutant lacking the entire N-terminal domain, but retaining the ZFs intact. Chromatin immunoprecipitation followed by sequencing was used to assess binding across the genome in murine embryonic fibroblasts. Unexpectedly, we observe that mutations in the N-terminal domain generally reduced binding, but there were also instances where binding was retained or even increased. These results provide a clear demonstration that the correct localization of TFs to their target genes is not solely dependent on their DNA-contact domains. This informs our understanding of how TFs operate and is of relevance to the design of artificial ZF proteins.


Asunto(s)
ADN/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular , Secuencia de Consenso , Regulación de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/química , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Mutación , Regiones Promotoras Genéticas , Unión Proteica , Estructura Terciaria de Proteína
12.
PLoS Genet ; 9(7): e1003612, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23874215

RESUMEN

KLF3 is a Krüppel family zinc finger transcription factor with widespread tissue expression and no previously known role in heart development. In a screen for dominant mutations affecting cardiovascular function in N-ethyl-N-nitrosourea (ENU) mutagenized mice, we identified a missense mutation in the Klf3 gene that caused aortic valvular stenosis and partially penetrant perinatal lethality in heterozygotes. All homozygotes died as embryos. In the first of three zinc fingers, a point mutation changed a highly conserved histidine at amino acid 275 to arginine (Klf3(H275R) ). This change impaired binding of the mutant protein to KLF3's canonical DNA binding sequence. Heterozygous Klf3(H275R) mutants that died as neonates had marked biventricular cardiac hypertrophy with diminished cardiac chambers. Adult survivors exhibited hypotension, cardiac hypertrophy with enlarged cardiac chambers, and aortic valvular stenosis. A dominant negative effect on protein function was inferred by the similarity in phenotype between heterozygous Klf3(H275R) mutants and homozygous Klf3 null mice. However, the existence of divergent traits suggested the involvement of additional interactions. We conclude that KLF3 plays diverse and important roles in cardiovascular development and function in mice, and that amino acid 275 is critical for normal KLF3 protein function. Future exploration of the KLF3 pathway provides a new avenue for investigating causative factors contributing to cardiovascular disorders in humans.


Asunto(s)
Estenosis de la Válvula Aórtica/genética , Enfermedades Cardiovasculares/genética , Factores de Transcripción de Tipo Kruppel/genética , Mutación Missense , Animales , Estenosis de la Válvula Aórtica/fisiopatología , Enfermedades Cardiovasculares/fisiopatología , Proteínas de Unión al ADN , Etilnitrosourea/química , Humanos , Factores de Transcripción de Tipo Kruppel/química , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Motivos de Nucleótidos/genética
13.
BMC Mol Biol ; 15: 8, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24885809

RESUMEN

BACKGROUND: Krüppel-like Factor 3 (KLF3) is a broadly expressed zinc-finger transcriptional repressor with diverse biological roles. During erythropoiesis, KLF3 acts as a feedback repressor of a set of genes that are activated by Krüppel-like Factor 1 (KLF1). Noting that KLF1 binds α-globin gene regulatory sequences during erythroid maturation, we sought to determine whether KLF3 also interacts with the α-globin locus to regulate transcription. RESULTS: We found that expression of a human transgenic α-globin reporter gene is markedly up-regulated in fetal and adult erythroid cells of Klf3-/- mice. Inspection of the mouse and human α-globin promoters revealed a number of canonical KLF-binding sites, and indeed, KLF3 was shown to bind to these regions both in vitro and in vivo. Despite these observations, we did not detect an increase in endogenous murine α-globin expression in Klf3-/- erythroid tissue. However, examination of murine embryonic fibroblasts lacking KLF3 revealed significant de-repression of α-globin gene expression. This suggests that KLF3 may contribute to the silencing of the α-globin locus in non-erythroid tissue. Moreover, ChIP-Seq analysis of murine fibroblasts demonstrated that across the locus, KLF3 does not occupy the promoter regions of the α-globin genes in these cells, but rather, binds to upstream, DNase hypersensitive regulatory regions. CONCLUSIONS: These findings reveal that the occupancy profile of KLF3 at the α-globin locus differs in erythroid and non-erythroid cells. In erythroid cells, KLF3 primarily binds to the promoters of the adult α-globin genes, but appears dispensable for normal transcriptional regulation. In non-erythroid cells, KLF3 distinctly binds to the HS-12 and HS-26 elements and plays a non-redundant, albeit modest, role in the silencing of α-globin expression.


Asunto(s)
Células Eritroides/metabolismo , Regulación de la Expresión Génica/genética , Factores de Transcripción de Tipo Kruppel/genética , Globinas alfa/genética , Animales , Sitios de Unión/genética , Células COS , Línea Celular Tumoral , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Células K562 , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Regiones Promotoras Genéticas/genética , Transcripción Genética/genética , Globinas alfa/metabolismo
14.
bioRxiv ; 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38370646

RESUMEN

Prior evidence indicates that the erythroid cellular response to glucocorticoids (GC) has developmental specificity, namely, that developmentally more advanced cells that are undergoing or have undergone fetal to adult globin switching are more responsive to GC-induced expansion. To investigate the molecular underpinnings of this, we focused on the major developmental globin regulator BCL11A. We compared: a) levels of expression and nuclear content of BCL11A in adult erythroid cells upon GC stimulation; b) response to GC of CD34+ cells from patients with BCL11A microdeletions and reduced BCL11A expression, and; c) response to GC of two cellular models (HUDEP-2 and adult CD34+ cells) before and after reduction of BCL11A expression by shRNA. We observed that: a) GC-expanded erythroid cells from a large cohort of blood donors displayed amplified expression and nuclear accumulation of BCL11A; b) CD34+ cells from BCL11A microdeletion patients generated fewer erythroid cells when cultured with GC compared to their parents, while the erythroid expansion of the patients was similar to that of their parents in cultures without GC, and; c) adult CD34+ cells and HUDEP-2 cells with shRNA-depleted expression of BCL11A exhibit reduced expansion in response to GC. In addition, RNA-seq profiling of shRNA-BCL11A CD34+ cells cultured with and without GC was similar (very few differentially expressed genes), while GC-specific responses (differential expression of GILZ and of numerous additional genes) were observed only in controls cells with unperturbed BCL11A expression. These data indicate that BCL11A is an important participant of certain aspects of the stress pathway sustained by GC.

15.
J Immunol ; 187(10): 5032-42, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22003205

RESUMEN

Krüppel-like factor 3 (Klf3) is a member of the Klf family of transcription factors. Klfs are widely expressed and have diverse roles in development and differentiation. In this study, we examine the function of Klf3 in B cell development by studying B lymphopoiesis in a Klf3 knockout mouse model. We show that B cell differentiation is significantly impaired in the bone marrow, spleen, and peritoneal cavity of Klf3 null mice and confirm that the defects are cell autonomous. In the bone marrow, there is a reduction in immature B cells, whereas recirculating mature cells are noticeably increased. Immunohistology of the spleen reveals a poorly structured marginal zone (MZ) that may in part be caused by deregulation of adhesion molecules on MZ B cells. In the peritoneal cavity, there are significant defects in B1 B cell development. We also report that the loss of Klf3 in MZ B cells is associated with reduced BCR signaling strength and an impaired ability to respond to LPS stimulation. Finally, we show increased expression of a number of Klf genes in Klf3 null B cells, suggesting that a Klf regulatory network may exist in B cells.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/patología , Factores de Transcripción de Tipo Kruppel/deficiencia , Factores de Transcripción de Tipo Kruppel/genética , Linfopoyesis/genética , Linfopoyesis/inmunología , Animales , Subgrupos de Linfocitos B/metabolismo , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peritoneo/inmunología , Peritoneo/metabolismo , Peritoneo/patología , Quimera por Radiación/genética , Quimera por Radiación/inmunología , Bazo/inmunología , Bazo/metabolismo , Bazo/patología
16.
J Biol Chem ; 286(44): 38190-38201, 2011 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-21908891

RESUMEN

Classical C2H2 zinc finger proteins are among the most abundant transcription factors found in eukaryotes, and the mechanisms through which they recognize their target genes have been extensively investigated. In general, a tandem array of three fingers separated by characteristic TGERP links is required for sequence-specific DNA recognition. Nevertheless, a significant number of zinc finger proteins do not contain a hallmark three-finger array of this type, raising the question of whether and how they contact DNA. We have examined the multi-finger protein ZNF217, which contains eight classical zinc fingers. ZNF217 is implicated as an oncogene and in repressing the E-cadherin gene. We show that two of its zinc fingers, 6 and 7, can mediate contacts with DNA. We examine its putative recognition site in the E-cadherin promoter and demonstrate that this is a suboptimal site. NMR analysis and mutagenesis is used to define the DNA binding surface of ZNF217, and we examine the specificity of the DNA binding activity using fluorescence anisotropy titrations. Finally, sequence analysis reveals that a variety of multi-finger proteins also contain two-finger units, and our data support the idea that these may constitute a distinct subclass of DNA recognition motif.


Asunto(s)
ADN/química , Transactivadores/fisiología , Secuencias de Aminoácidos , Sitios de Unión , Núcleo Celular/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Espectroscopía de Resonancia Magnética/métodos , Modelos Moleculares , Unión Proteica , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Transactivadores/química , Transcripción Genética , Dedos de Zinc
17.
Adv Exp Med Biol ; 747: 105-21, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22949114

RESUMEN

Eukaryotic transcription factors bind DNA and typically serve to localize large multiprotein complexes to particular genes to up- or downregulate transcription, thereby coordinating cellular responses to a variety of signals. Different combinations of transcription factors within DNA-binding multiprotein complexes allow individual proteins to partake in multiple different regulatory pathways. Many transcription factors can form homo- and heterodimers (or oligomers) with different partners, thus modulating DNA-binding specificity and affinity and/or the recruitment of different binding partners. This chapter reviews several of the mechanisms by which the homo- and heterodimerization of transcription factors contributes to transcriptional regulation.


Asunto(s)
ADN/genética , Multimerización de Proteína , Factores de Transcripción/genética , Transcripción Genética , Animales , Sitios de Unión , ADN/química , ADN/metabolismo , Dimerización , Regulación de la Expresión Génica , Humanos , Modelos Moleculares , Unión Proteica , Factores de Transcripción/química , Factores de Transcripción/metabolismo
18.
IUBMB Life ; 63(2): 86-93, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21360637

RESUMEN

KLF3 is a member of the Krüppel-like factor (KLF) family of transcription factors. These proteins are classified by the presence of three C-terminal C2H2 zinc fingers that allow sequence-specific binding to CACCC boxes and GC-rich motifs found in the promoters, enhancers, and other control regions of target genes. KLFs have diverse biological roles, regulating proliferation, differentiation, and apoptosis in many tissues throughout development. KLF3 is a transcriptional repressor that binds the cofactor C-terminal binding protein, which in turn recruits a large repressor complex to mediate transcriptional silencing. In addition to an understanding of the molecular mechanisms that allow KLF3 to regulate the expression of its target genes, the biological roles of this transcription factor are now being defined. In agreement with the widespread expression pattern of this transcription factor, it is becoming clear that KLF3 is an important regulator of several biological processes, including adipogenesis, erythropoiesis, and B cell development.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/metabolismo , Regiones Promotoras Genéticas , Proteínas Represoras/metabolismo , Transcripción Genética , Adipogénesis/genética , Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/metabolismo , Secuencia de Aminoácidos , Animales , Apoptosis , Linfocitos B/fisiología , Diferenciación Celular/genética , Línea Celular , Proliferación Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Eritropoyesis , Secuencia Rica en GC , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Mamíferos/genética , Mamíferos/metabolismo , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Filogenia , Proteínas Represoras/genética , Zinc/metabolismo , Dedos de Zinc/genética
19.
Mol Cell Biol ; 27(7): 2777-90, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17283065

RESUMEN

The Sp/Krüppel-like factor (Sp/Klf) family is comprised of around 25 zinc finger transcription factors that recognize CACCC boxes and GC-rich elements. We have investigated basic Krüppel-like factor (Bklf/Klf3) and show that in erythroid tissues its expression is highly dependent on another family member, erythroid Krüppel-like factor (Eklf/Klf1). We observe that Bklf mRNA is significantly reduced in erythroid tissues from Eklf-null murine embryos. We find that Bklf is driven primarily by two promoters, a ubiquitously active GC-rich upstream promoter, 1a, and an erythroid downstream promoter, 1b. Transcripts from the two promoters encode identical proteins. Interestingly, both the ubiquitous and the erythroid promoter are dependent on Eklf in erythroid cells. Eklf also activates both promoters in transient assays. Experiments utilizing an inducible form of Eklf demonstrate activation of the endogenous Bklf gene in the presence of an inhibitor of protein synthesis. The kinetics of activation are also consistent with Bklf being a direct Eklf target. Chromatin immunoprecipitation assays confirm that Eklf associates with both Bklf promoters. Eklf is typically an activator of transcription, whereas Bklf is noted as a repressor. Our results support the hypothesis that feedback cross-regulation occurs within the Sp/Klf family in vivo.


Asunto(s)
Células Eritroides/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/fisiología , Animales , Secuencia de Bases , Feto/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Hígado/crecimiento & desarrollo , Hígado/metabolismo , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Regiones Promotoras Genéticas
20.
Cell Rep ; 31(8): 107676, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32460018

RESUMEN

The human genome encodes millions of regulatory elements, of which only a small fraction are active within a given cell type. Little is known about the global impact of chromatin remodelers on regulatory DNA landscapes and how this translates to gene expression. We use precision genome engineering to reawaken homozygously inactivated SMARCA4, a central ATPase of the human SWI/SNF chromatin remodeling complex, in lung adenocarcinoma cells. Here, we combine DNase I hypersensitivity, histone modification, and transcriptional profiling to show that SMARCA4 dramatically increases both the number and magnitude of accessible chromatin sites genome-wide, chiefly by unmasking sites of low regulatory factor occupancy. By contrast, transcriptional changes are concentrated within well-demarcated remodeling domains wherein expression of specific genes is gated by both distal element activation and promoter chromatin configuration. Our results provide a perspective on how global chromatin remodeling activity is translated to gene expression via regulatory DNA.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , ADN Helicasas/metabolismo , ADN/genética , Expresión Génica/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA