Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
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
2.
Mol Ther ; 24(8): 1351-7, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27434588

RESUMEN

Cystic fibrosis (CF) is a life-shortening genetic disease. The root cause of CF is heritable recessive mutations that affect the cystic fibrosis transmembrance conductance regulator (CFTR) gene and the subsequent expression and activity of encoded ion channels at the cell surface. We show that CFTR is regulated transcriptionally by the actions of a novel long noncoding RNA (lncRNA), designated as BGas, that emanates from intron 11 of the CFTR gene and is expressed in the antisense orientation relative to the protein coding sense strand. We find that BGas functions in concert with several proteins including HMGA1, HMGB1, and WIBG to modulate the local chromatin and DNA architecture of intron 11 of the CFTR gene and thereby affects transcription. Suppression of BGas or its associated proteins results in a gain of both CFTR expression and chloride ion function. The observations described here highlight a previously underappreciated mechanism of transcriptional control and suggest that BGas may serve as a therapeutic target for specifically activating expression of CFTR.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/genética , Regulación de la Expresión Génica , ARN sin Sentido/genética , ARN Largo no Codificante , Fibrosis Quística/metabolismo , Proteínas de Unión al ADN/metabolismo , Sitios Genéticos , Humanos , Modelos Biológicos , Unión Proteica
3.
Trends Biochem Sci ; 37(5): 199-205, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22405571

RESUMEN

Classical zinc fingers (ZFs) are one of the most common protein domains in higher eukaryotes and have been known for almost 30 years to act as sequence-specific DNA-binding domains. This knowledge has come, however, from the study of a small number of archetypal proteins, and a larger picture is beginning to emerge that ZF functions are far more diverse than originally suspected. Here, we review the evidence that a subset of ZF proteins live double lives, binding to both DNA and RNA targets and frequenting both the cytoplasm and the nucleus. This duality can create an important additional level of gene regulation that serves to connect transcriptional and post-transcriptional control.


Asunto(s)
Proteínas de Unión al ARN/metabolismo , ARN/metabolismo , Factores de Transcripción/metabolismo , Dedos de Zinc , ADN/química , ADN/genética , ADN/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Humanos , Modelos Moleculares , Unión Proteica , ARN/química , ARN/genética , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Factores de Transcripción/química , Factores de Transcripción/genética
4.
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
5.
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
6.
RNA Biol ; 12(8): 893-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26156430

RESUMEN

Amplification or overexpression of neuronal MYC (MYCN) is associated with poor prognosis of human neuroblastoma. Three isoforms of the MYCN protein have been described as well as a protein encoded by an antisense transcript (MYCNOS) that originates from the opposite strand at the MYCN locus. Recent findings suggest that some antisense long non-coding RNAs (lncRNAs) can play a role in epigenetically regulating gene expression. Here we report that MYCNOS transcripts function as a modulator of the MYCN locus, affecting MYCN promoter usage and recruiting various proteins, including the Ras GTPase-activating protein-binding protein G3BP1, to the upstream MYCN promoter. Overexpression of MYCNOS results in a reduction of upstream MYCN promoter usage and increased MYCN expression, suggesting that the protein-coding MYCNOS also functions as a regulator of MYCN ultimately controlling MYCN transcriptional variants. The observations presented here demonstrate that protein-coding transcripts can regulate gene transcription and can tether regulatory proteins to target loci.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Proteínas Nucleares/genética , Proteínas Oncogénicas/genética , Regiones Promotoras Genéticas/genética , ARN sin Sentido/genética , Azacitidina/farmacología , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , ADN Helicasas , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ácidos Hidroxámicos/farmacología , Modelos Genéticos , Proteína Proto-Oncogénica N-Myc , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Proteínas de Unión a Poli-ADP-Ribosa , Isoformas de Proteínas/genética , Inhibidores de la Síntesis de la Proteína/farmacología , ARN Helicasas , Proteínas con Motivos de Reconocimiento de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
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
8.
Nat Genet ; 50(4): 498-503, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29610478

RESUMEN

ß-hemoglobinopathies such as sickle cell disease (SCD) and ß-thalassemia result from mutations in the adult HBB (ß-globin) gene. Reactivating the developmentally silenced fetal HBG1 and HBG2 (γ-globin) genes is a therapeutic goal for treating SCD and ß-thalassemia 1 . Some forms of hereditary persistence of fetal hemoglobin (HPFH), a rare benign condition in which individuals express the γ-globin gene throughout adulthood, are caused by point mutations in the γ-globin gene promoter at regions residing ~115 and 200 bp upstream of the transcription start site. We found that the major fetal globin gene repressors BCL11A and ZBTB7A (also known as LRF) directly bound to the sites at -115 and -200 bp, respectively. Furthermore, introduction of naturally occurring HPFH-associated mutations into erythroid cells by CRISPR-Cas9 disrupted repressor binding and raised γ-globin gene expression. These findings clarify how these HPFH-associated mutations operate and demonstrate that BCL11A and ZBTB7A are major direct repressors of the fetal globin gene.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Unión al ADN/metabolismo , Hemoglobina Fetal/genética , Mutación , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , gamma-Globinas/genética , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Secuencia de Bases , Sitios de Unión/genética , Sistemas CRISPR-Cas , Línea Celular , Humanos , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/metabolismo , Sitio de Iniciación de la Transcripción , Talasemia beta/genética , Talasemia beta/terapia
9.
Blood Adv ; 1(11): 685-692, 2017 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-29296711

RESUMEN

Genes encoding the human ß-like hemoglobin proteins undergo a developmental switch from fetal γ-globin to adult ß-globin expression around the time of birth. ß-hemoglobinopathies, such as sickle-cell disease and ß-thalassemia, result from mutations affecting the adult ß-globin gene. The only treatment options currently available carry significant adverse effects. Analyses of heritable variations in fetal hemoglobin (HbF) levels have provided evidence that reactivation of the silenced fetal γ-globin genes in adult erythroid cells is a promising therapy. The γ-globin repressor BCL11A has become the major focus, with several studies investigating its regulation and function as a first step to inhibiting its expression or activity. However, a second repression mechanism was recently shown to be mediated by the transcription factor ZBTB7A/LRF, suggesting that understanding the regulation of ZBTB7A may also be useful. Here we show that Krüppel-like factor 1 (KLF1) directly drives expression of ZBTB7A in erythroid cells by binding to its proximal promoter. We have also uncovered an erythroid-specific regulation mechanism, leading to the upregulation of a novel ZBTB7A transcript in the erythroid compartment. The demonstration that ZBTB7A, like BCL11A, is a KLF1 target gene also fits with the observation that reduced KLF1 expression or activity is associated with HbF derepression.

10.
PLoS One ; 11(4): e0152424, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27082860

RESUMEN

Natural selective processes have been known to drive phenotypic plasticity, which is the emergence of different phenotypes from one genome following environmental stimulation. Long non-coding RNAs (lncRNAs) have been observed to modulate transcriptional and epigenetic states of genes in human cells. We surmised that lncRNAs are governors of phenotypic plasticity and drive natural selective processes through epigenetic modulation of gene expression. Using heat shocked human cells as a model we find several differentially expressed transcripts with the top candidates being lncRNAs derived from retro-elements. One particular retro-element derived transcripts, Retro-EIF2S2, was found to be abundantly over-expressed in heat shocked cells. Over-expression of Retro-EIF2S2 significantly enhanced cell viability and modulated a predisposition for an adherent cellular phenotype upon heat shock. Mechanistically, we find that this retro-element derived transcript interacts directly with a network of proteins including 40S ribosomal protein S30 (FAU), Eukaryotic translation initiation factor 5A (EIF5A), and Ubiquitin-60S ribosomal protein L40 (UBA52) to affect protein modulated cell adhesion pathways. We find one motif in Retro-EIF2S2 that exhibits binding to FAU and modulates phenotypic cell transitions from adherent to suspension states. The observations presented here suggest that retroviral derived transcripts actively modulate phenotypic plasticity in human cells in response to environmental selective pressures and suggest that natural selection may play out through the action of retro-elements in human cells.


Asunto(s)
Plasticidad de la Célula/genética , Epigénesis Genética/genética , Regulación de la Expresión Génica , Respuesta al Choque Térmico/genética , ARN Largo no Codificante/genética , Células HEK293 , Humanos , Fenotipo , Retroelementos/genética
11.
Noncoding RNA Res ; 1(1): 3-11, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28090596

RESUMEN

Cells communicate with one another to create microenvironments and share resources. One avenue by which cells communicate is through the action of exosomes. Exosomes are extracellular vesicles that are released by one cell and taken up by neighbouring cells. But how exosomes instigate communication between cells has remained largely unknown. We present evidence here that particular long non-coding RNA molecules are preferentially packaged into exosomes. We also find that a specific class of these exosome associated non-coding RNAs functionally modulate cell viability by direct interactions with L-lactate dehydrogenase B (LDHB), high-mobility group protein 17 (HMG-17), and CSF2RB, proteins involved in metabolism, nucleosomal architecture and cell signalling respectively. Knowledge of this endogenous cell to cell pathway, those proteins interacting with exosome associated non-coding transcripts and their interacting domains, could lead to a better understanding of not only cell to cell interactions but also the development of exosome targeted approaches in patient specific cell-based therapies.

12.
Australas J Ultrasound Med ; 18(3): 96-99, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28191249

RESUMEN

Infection control and prevention is critical to delivering safe and high-quality care to patients undergoing sonographic procedures. In Australia comprehensive standards for reprocessing of ultrasound probes are based on the AS/NZS, TGA and ASUM recommendations. These standards align with the US Centers for Disease Control and Prevention recommendations. However compliance to these guidelines is not ideal and there exists an unmet need for refinement of the guidelines relating to specific factors in clinical sonography. Significant microbiological evidence exists reflecting the increased risk of infection transmission specifically through inadequately reprocessed ultrasound probes. Studies have reported > 80% of transvaginal ultrasound probe handles are contaminated with disease causing pathogens since handle disinfection is omitted from standard reprocessing protocols. Significantly, it was recently discovered that widely-used high level disinfectants referred to in guidelines are unable to kill HPV while it is becoming increasingly apparent that attention must be paid to the clinical sonography environment as a potential source of nosocomial pathogens. Ultrasound probe reprocessing guidelines and standards are comprehensive however the challenge is in general awareness and effective implementation into practice. As future research in this area is performed, guidelines will need to be amenable to revision to provide patients with the best standard of care.

13.
Infect Control Hosp Epidemiol ; 36(5): 581-4, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25723900

RESUMEN

Intracavity ultrasound transducer handles are not routinely immersed in liquid high-level disinfectants. We show that residual bacteria, including pathogens, persist on more than 80% of handles that are not disinfected, whereas use of an automated device reduces contamination to background levels. Clinical staff should consider the need for handle disinfection.


Asunto(s)
Infección Hospitalaria/prevención & control , Desinfección/métodos , Endosonografía/efectos adversos , Contaminación de Equipos/prevención & control , Ultrasonografía Prenatal/efectos adversos , Infección Hospitalaria/etiología , Estudios Transversales , Endosonografía/normas , Femenino , Humanos , Embarazo , Factores de Riesgo , Ultrasonografía Prenatal/métodos
14.
J Infect Public Health ; 7(2): 153-60, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24314741

RESUMEN

BACKGROUND: Ultrasound transducer reprocessing is required to prevent the transmission of infections between patients. In some regions, reprocessing practices are not sufficient to achieve high-level disinfection (HLD), which can result in contaminated probes. Furthermore, current manual HLD methods use toxic chemicals and are prone to operator error/variability. The development of automated, non-toxic HLD disinfection devices may reduce the risk of transmission and reduce safety risks for operators and patients. This study investigated the disinfection efficacy of a hydrogen peroxide-based, automated HLD device, the Trophon(®) EPR, against a range of international standards. METHODS: Disinfection efficacy was assessed in carrier and simulated use tests against 21 different species of bacteria, fungi and viruses. Carrier tests were performed by placing carriers throughout the disinfection chamber and measuring the log reduction in viable organisms following disinfection. These tests were performed according to Association of Analytical Communities International Official Methods and European and ASTM International Standards for bactericidal, fungicidal, mycobactericidal, sporicidal and virucidal disinfection. Simulated use tests involving the disinfection of six widely used ultrasound probe models were conducted according to ASTM-E1837-96 using Mycobacterium terrae as a test organism. RESULTS: The device satisfied criteria for HLD and sporicidal disinfection efficacy under all standards tested. CONCLUSIONS: Automated, hydrogen peroxide-based disinfection devices offer an alternative to manual ultrasound probe disinfection technologies. Such devices reduce the risks of operator error and can improve patient and operator safety by preventing exposure to toxic chemicals. The adoption of next-generation disinfection devices may help to decrease infection risk and improve patient safety.


Asunto(s)
Desinfectantes/farmacología , Desinfección/métodos , Equipos y Suministros/microbiología , Equipos y Suministros/virología , Peróxido de Hidrógeno/farmacología , Bacterias/efectos de los fármacos , Hongos/efectos de los fármacos , Viabilidad Microbiana/efectos de los fármacos , Ultrasonografía/instrumentación , Virus/efectos de los fármacos
15.
Genome Biol ; 15(4): R58, 2014 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-24946810

RESUMEN

BACKGROUND: Retroviral elements are pervasively transcribed and dynamically regulated during development. While multiple histone- and DNA-modifying enzymes have broadly been associated with their global silencing, little is known about how the many diverse retroviral families are each selectively recognized. RESULTS: Here we show that the zinc finger protein Krüppel-like Factor 3 (KLF3) specifically silences transcription from the ORR1A0 long terminal repeat in murine fetal and adult erythroid cells. In the absence of KLF3, we detect widespread transcription from ORR1A0 elements driven by the master erythroid regulator KLF1. In several instances these aberrant transcripts are spliced to downstream genic exons. One such chimeric transcript produces a novel, dominant negative isoform of PU.1 that can induce erythroid differentiation. CONCLUSIONS: We propose that KLF3 ensures the integrity of the murine erythroid transcriptome through the selective repression of a particular retroelement and is likely one of multiple sequence-specific factors that cooperate to achieve global silencing.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/metabolismo , Retroelementos/genética , Secuencias Repetidas Terminales/genética , Animales , Línea Celular , Células Eritroides/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcriptoma
16.
Diabetes ; 62(8): 2728-37, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23633521

RESUMEN

Krüppel-like factor 3 (KLF3) is a transcriptional regulator that we have shown to be involved in the regulation of adipogenesis in vitro. Here, we report that KLF3-null mice are lean and protected from diet-induced obesity and glucose intolerance. On a chow diet, plasma levels of leptin are decreased, and adiponectin is increased. Despite significant reductions in body weight and adiposity, wild-type and knockout animals show equivalent energy intake, expenditure, and excretion. To investigate the molecular events underlying these observations, we used microarray analysis to compare gene expression in Klf3(+/+) and Klf3(-/-) tissues. We found that mRNA expression of Fam132a, which encodes a newly identified insulin-sensitizing adipokine, adipolin, is significantly upregulated in the absence of KLF3. We confirmed that KLF3 binds the Fam132a promoter in vitro and in vivo and that this leads to repression of promoter activity. Further, plasma adipolin levels were significantly increased in Klf3(-/-) mice compared with wild-type littermates. Boosting levels of adipolin via targeting of KLF3 offers a novel potential therapeutic strategy for the treatment of insulin resistance.


Asunto(s)
Adipoquinas/genética , Regulación de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/genética , Regulación hacia Arriba/genética , Adipoquinas/sangre , Adipoquinas/metabolismo , Animales , Metabolismo Energético/fisiología , Factores de Transcripción de Tipo Kruppel/sangre , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas
17.
Mol Cell Biol ; 32(16): 3281-92, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22711990

RESUMEN

The CACCC-box binding protein erythroid Krüppel-like factor (EKLF/KLF1) is a master regulator that directs the expression of many important erythroid genes. We have previously shown that EKLF drives transcription of the gene for a second KLF, basic Krüppel-like factor, or KLF3. We have now tested the in vivo role of KLF3 in erythroid cells by examining Klf3 knockout mice. KLF3-deficient adults exhibit a mild compensated anemia, including enlarged spleens, increased red pulp, and a higher percentage of erythroid progenitors, together with elevated reticulocytes and abnormal erythrocytes in the peripheral blood. Impaired erythroid maturation is also observed in the fetal liver. We have found that KLF3 levels rise as erythroid cells mature to become TER119(+). Consistent with this, microarray analysis of both TER119(-) and TER119(+) erythroid populations revealed that KLF3 is most critical at the later stages of erythroid maturation and is indeed primarily a transcriptional repressor. Notably, many of the genes repressed by KLF3 are also known to be activated by EKLF. However, the majority of these are not currently recognized as erythroid-cell-specific genes. These results reveal the molecular and physiological function of KLF3, defining it as a feedback repressor that counters the activity of EKLF at selected target genes to achieve normal erythropoiesis.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/metabolismo , Animales , Antígenos de Grupos Sanguíneos/genética , Inmunoprecipitación de Cromatina , Eritrocitos/citología , Eritropoyesis , Citometría de Flujo/métodos , Perfilación de la Expresión Génica , Ratones , Ratones Noqueados , Modelos Genéticos , Análisis de Secuencia por Matrices de Oligonucleótidos , Bazo/citología , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA