Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
mBio ; 12(6): e0268421, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34749533

RESUMEN

The life cycle of human papillomavirus (HPV) depends on keratinocyte differentiation as the virus modulates and takes advantage of cellular pathways to replicate its genome and assemble viral particles in differentiated cells. Viral genomes are amplified in nuclear replication foci in differentiated keratinocytes, and DNA repair factors from the DNA damage response signaling pathway are recruited to replicate viral DNA. The HPV genome is associated with cellular histones at all stages of the infectious cycle, and here, we show that the histone variant macroH2A1 is bound to the HPV genome and enriched in viral replication foci in differentiated cells. macroH2A1 isoforms play important roles in cellular transcriptional repression, double-strand break repair, and replication stress. The viral E8^E2 protein also binds to the HPV genome and inhibits viral replication and gene expression by recruiting NCoR/SMRT complexes. We show here that E8^E2 and SMRT also localize within replication foci, though independently from macroH2A1. Conversely, transcription complexes containing RNA polymerase II and Brd4 are located on the surface of the foci. Foci generated with an HPV16 E8^E2 mutant genome are not enriched for SMRT or macroH2A1 but contain transcriptional complexes throughout the foci. We propose that both the cellular macroH2A1 protein and viral E8^E2 protein help to spatially separate replication and transcription activities within viral replication foci. IMPORTANCE Human papillomaviruses are small DNA viruses that cause chronic infection of cutaneous and mucosal epithelium. In some cases, persistent infection with HPV can result in cancer, and 5% of human cancers are the result of HPV infection. In differentiated cells, HPV amplifies viral DNA in nuclear replication factories and transcribes late mRNAs to produce capsid proteins. However, very little is known about the spatial organization of these activities in the nucleus. Here, we show that repressive viral and cellular factors localize within the foci to suppress viral transcription, while active transcription takes place on the surface. The cellular histone variant macroH2A1 is important for this spatial organization.


Asunto(s)
Alphapapillomavirus/fisiología , Genoma Viral , Infecciones por Papillomavirus/virología , Replicación Viral , Alphapapillomavirus/genética , Histonas/genética , Histonas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Co-Represor 1 de Receptor Nuclear/genética , Co-Represor 1 de Receptor Nuclear/metabolismo , Co-Represor 2 de Receptor Nuclear/genética , Co-Represor 2 de Receptor Nuclear/metabolismo , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/metabolismo , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/metabolismo
2.
Front Mol Biosci ; 8: 725794, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34386523

RESUMEN

Human Papillomaviruses (HPV) reproduce in stratified epithelia by establishing a reservoir of low- level infection in the dividing basal cells and restricting the production of viral particles to terminally differentiated cells. These small DNA viruses hijack pivotal cellular processes and pathways to support the persistent infectious cycle. One cellular factor that is key to multiple stages of viral replication and transcription is the BET (bromodomain and extra-terminal domain) protein, Brd4 (Bromodomain containing protein 4). Here we provide an overview of the multiple interactions of Brd4 that occur throughout the HPV infectious cycle.

3.
Viruses ; 13(2)2021 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-33672465

RESUMEN

Persistent infection with oncogenic human papillomavirus (HPV) types is responsible for ~5% of human cancers. The HPV infectious cycle can sustain long-term infection in stratified epithelia because viral DNA is maintained as low copy number extrachromosomal plasmids in the dividing basal cells of a lesion, while progeny viral genomes are amplified to large numbers in differentiated superficial cells. The viral E1 and E2 proteins initiate viral DNA replication and maintain and partition viral genomes, in concert with the cellular replication machinery. Additionally, the E5, E6, and E7 proteins are required to evade host immune responses and to produce a cellular environment that supports viral DNA replication. An unfortunate consequence of the manipulation of cellular proliferation and differentiation is that cells become at high risk for carcinogenesis.


Asunto(s)
Papillomaviridae/fisiología , Infecciones por Papillomavirus/virología , Animales , ADN Viral/genética , Genoma Viral , Humanos , Papillomaviridae/genética , Replicación Viral
4.
Mol Cell Oncol ; 5(3): e1441629, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30250894

RESUMEN

The macroH2A1.2 histone variant facilitates the response to replication stress with implications for genome maintenance and cell growth. A mutually exclusive splice variant, macroH2A1.1, has opposing effects on DNA repair outcome and proliferation. Here we discuss the potential impact of splicing-modulated macroH2A1 chromatin organization for cell function and malignant transformation.

5.
Mol Cell ; 69(1): 36-47.e7, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29249653

RESUMEN

Recent integrative epigenome analyses highlight the importance of functionally distinct chromatin states for accurate cell function. How these states are established and maintained is a matter of intense investigation. Here, we present evidence for DNA damage as an unexpected means to shape a protective chromatin environment at regions of recurrent replication stress (RS). Upon aberrant fork stalling, DNA damage signaling and concomitant H2AX phosphorylation coordinate the FACT-dependent deposition of macroH2A1.2, a histone variant that promotes DNA repair by homologous recombination (HR). MacroH2A1.2, in turn, facilitates the accumulation of the tumor suppressor and HR effector BRCA1 at replication forks to protect from RS-induced DNA damage. Consequently, replicating primary cells steadily accrue macroH2A1.2 at fragile regions, whereas macroH2A1.2 loss in these cells triggers DNA damage signaling-dependent senescence, a hallmark of RS. Altogether, our findings demonstrate that recurrent DNA damage contributes to the chromatin landscape to ensure the epigenomic integrity of dividing cells.


Asunto(s)
Carcinogénesis/genética , Cromatina/genética , Daño del ADN/genética , Reparación del ADN/genética , Replicación del ADN/genética , Histonas/genética , Recombinación Homóloga/genética , Proteína BRCA1/metabolismo , División Celular/genética , Células Cultivadas , Senescencia Celular/genética , Inestabilidad Genómica/fisiología , Humanos , Transducción de Señal/genética
6.
PLoS Pathog ; 13(10): e1006660, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28968443

RESUMEN

We have shown previously that Sp100 (a component of the ND10 nuclear body) represses transcription, replication and establishment of incoming human papillomavirus (HPV) DNA in the early stages of infection. In this follow up study, we show that Sp100 does not substantially regulate viral infection in the maintenance phase, however at late stages of infection Sp100 interacts with amplifying viral genomes to repress viral processes. We find that Sp100 localizes to HPV16 replication foci generated in primary keratinocytes, to HPV31 replication foci that form in differentiated cells, and to HPV16 replication foci in CIN 1 cervical biopsies. To analyze this further, Sp100 was down regulated by siRNA treatment of differentiating HPV31 containing cells and levels of viral transcription and replication were assessed. This revealed that Sp100 represses viral transcription and replication in differentiated cells. Analysis of Sp100 binding to viral chromatin showed that Sp100 bound across the viral genome, and that binding increased at late stages of infection. Therefore, Sp100 represses the HPV life cycle at both early and late stages of infection.


Asunto(s)
Antígenos Nucleares/metabolismo , Autoantígenos/metabolismo , Replicación del ADN/genética , Papillomavirus Humano 16/metabolismo , Queratinocitos/metabolismo , Replicación Viral , Genoma Viral , Interacciones Huésped-Patógeno/inmunología , Papillomavirus Humano 16/genética , Humanos , Queratinocitos/inmunología , Factores de Transcripción/metabolismo , Replicación Viral/genética
7.
J Biol Chem ; 292(5): 1637-1647, 2017 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-27998979

RESUMEN

Glucocorticoids are a general class of steroids that possess renoprotective activity in glomeruli through their interaction with the glucocorticoid receptor. However, the mechanisms by which glucocorticoids ameliorate proteinuria and glomerular disease are not well understood. In this study, we demonstrated that α actinin 4 (ACTN4), an actin-cross-linking protein known to coordinate cytoskeletal organization, interacts with the glucocorticoid receptor (GR) in the nucleus of human podocytes (HPCs), a key cell type in the glomerulus critical for kidney filtration function. The GR-ACTN4 complex enhances glucocorticoid response element (GRE)-driven reporter activity. Stable knockdown of ACTN4 by shRNA in HPCs significantly reduces dexamethasone-mediated induction of GR target genes and GRE-driven reporter activity without disrupting dexamethasone-induced nuclear translocation of GR. Synonymous mutations or protein expression losses in ACTN4 are associated with kidney diseases, including focal segmental glomerulosclerosis, characterized by proteinuria and podocyte injury. We found that focal segmental glomerulosclerosis-linked ACTN4 mutants lose their ability to bind liganded GR and support GRE-mediated transcriptional activity. Mechanistically, GR and ACTN4 interact in the nucleus of HPCs. Furthermore, disruption of the LXXLL nuclear receptor-interacting motif present in ACTN4 results in reduced GR interaction and dexamethasone-mediated transactivation of a GRE reporter while still maintaining its actin-binding activity. In contrast, an ACTN4 isoform, ACTN4 (Iso), that loses its actin-binding domain is still capable of potentiating a GRE reporter. Dexamethasone induces the recruitment of ACTN4 and GR to putative GREs in dexamethasone-transactivated promoters, SERPINE1, ANGPLT4, CCL20, and SAA1 as well as the NF-κB (p65) binding sites on GR-transrepressed promoters such as IL-1ß, IL-6, and IL-8 Taken together, our data establish ACTN4 as a transcriptional co-regulator that modulates both dexamethasone-transactivated and -transrepressed genes in podocytes.


Asunto(s)
Actinina/biosíntesis , Dexametasona/farmacología , Podocitos/metabolismo , Receptores de Glucocorticoides/metabolismo , Elementos de Respuesta/fisiología , Activación Transcripcional/efectos de los fármacos , Actinina/genética , Citocinas/biosíntesis , Citocinas/genética , Células HEK293 , Células HeLa , Humanos , Podocitos/citología , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Receptores de Glucocorticoides/agonistas , Receptores de Glucocorticoides/genética
8.
Genes (Basel) ; 6(3): 858-77, 2015 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-26378584

RESUMEN

DNA replication is essential for cell division. Challenges to the progression of DNA polymerase can result in replication stress, promoting the stalling and ultimately collapse of replication forks. The latter involves the formation of DNA double-strand breaks (DSBs) and has been linked to both genome instability and irreversible cell cycle arrest (senescence). Recent technological advances have elucidated many of the factors that contribute to the sensing and repair of stalled or broken replication forks. In addition to bona fide repair factors, these efforts highlight a range of chromatin-associated changes at and near sites of replication stress, suggesting defects in epigenome maintenance as a potential outcome of aberrant DNA replication. Here, we will summarize recent insight into replication stress-induced chromatin-reorganization and will speculate on possible adverse effects for gene expression, nuclear integrity and, ultimately, cell function.

9.
EMBO Rep ; 16(11): 1520-34, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26412854

RESUMEN

Long non-coding RNAs (lncRNAs) are important players in diverse biological processes. Upon DNA damage, cells activate a complex signaling cascade referred to as the DNA damage response (DDR). Using a microarray screen, we identify here a novel lncRNA, DDSR1 (DNA damage-sensitive RNA1), which is induced upon DNA damage. DDSR1 induction is triggered in an ATM-NF-κB pathway-dependent manner by several DNA double-strand break (DSB) agents. Loss of DDSR1 impairs cell proliferation and DDR signaling and reduces DNA repair capacity by homologous recombination (HR). The HR defect in the absence of DDSR1 is marked by aberrant accumulation of BRCA1 and RAP80 at DSB sites. In line with a role in regulating HR, DDSR1 interacts with BRCA1 and hnRNPUL1, an RNA-binding protein involved in DNA end resection. Our results suggest a role for the lncRNA DDSR1 in modulating DNA repair by HR.


Asunto(s)
Proteína BRCA1/metabolismo , Daño del ADN , Recombinación Homóloga , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Proliferación Celular , Roturas del ADN de Doble Cadena , Reparación del ADN , Regulación de la Expresión Génica , Genes BRCA1 , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Análisis por Micromatrices , FN-kappa B/metabolismo , Proteínas Nucleares/metabolismo , ARN Largo no Codificante/aislamiento & purificación , Transducción de Señal , Factores de Transcripción/metabolismo
10.
Cell Rep ; 8(4): 1049-62, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25131201

RESUMEN

Appropriate DNA double-strand break (DSB) repair factor choice is essential for ensuring accurate repair outcome and genomic integrity. The factors that regulate this process remain poorly understood. Here, we identify two repressive chromatin components, the macrohistone variant macroH2A1 and the H3K9 methyltransferase and tumor suppressor PRDM2, which together direct the choice between the antagonistic DSB repair mediators BRCA1 and 53BP1. The macroH2A1/PRDM2 module mediates an unexpected shift from accessible to condensed chromatin that requires the ataxia telangiectasia mutated (ATM)-dependent accumulation of both proteins at DSBs in order to promote DSB-flanking H3K9 dimethylation. Remarkably, loss of macroH2A1 or PRDM2, as well as experimentally induced chromatin decondensation, impairs the retention of BRCA1, but not 53BP1, at DSBs. As a result, macroH2A1 and/or PRDM2 depletion causes epistatic defects in DSB end resection, homology-directed repair, and the resistance to poly(ADP-ribose) polymerase (PARP) inhibition-all hallmarks of BRCA1-deficient tumors. Together, these findings identify dynamic, DSB-associated chromatin reorganization as a critical modulator of BRCA1-dependent genome maintenance.


Asunto(s)
Proteína BRCA1/fisiología , Histonas/fisiología , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/metabolismo , Inestabilidad Genómica , Células HEK293 , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Metilación , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/fisiología , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Reparación del ADN por Recombinación , Factores de Transcripción/metabolismo
11.
PLoS One ; 8(4): e60213, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23593176

RESUMEN

Glomerular podocytes are highly differentiated epithelial cells that are key components of the kidney filtration units. Podocyte damage or loss is the hallmark of nephritic diseases characterized by severe proteinuria. Recent studies implicate that hormones including glucocorticoids (ligand for glucocorticoid receptor) and vitamin D3 (ligand for vitamin D receptor) protect or promote repair of podocytes from injury. In order to elucidate the mechanisms underlying hormone-mediated podocyte-protecting activity from injury, we carried out microarray gene expression studies to identify the target genes and corresponding pathways in response to these hormones during podocyte differentiation. We used immortalized human cultured podocytes (HPCs) as a model system and carried out in vitro differentiation assays followed by dexamethasone (Dex) or vitamin D3 (VD3) treatment. Upon the induction of differentiation, multiple functional categories including cell cycle, organelle dynamics, mitochondrion, apoptosis and cytoskeleton organization were among the most significantly affected. Interestingly, while Dex and VD3 are capable of protecting podocytes from injury, they only share limited target genes and affected pathways. Compared to VD3 treatment, Dex had a broader and greater impact on gene expression profiles. In-depth analyses of Dex altered genes indicate that Dex crosstalks with a broad spectrum of signaling pathways, of which inflammatory responses, cell migration, angiogenesis, NF-κB and TGFß pathways are predominantly altered. Together, our study provides new information and identifies several new avenues for future investigation of hormone signaling in podocytes.


Asunto(s)
Colecalciferol/farmacología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Glucocorticoides/farmacología , Podocitos/efectos de los fármacos , Podocitos/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular Tumoral , Células Cultivadas , Análisis por Conglomerados , Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , Humanos , Anotación de Secuencia Molecular , Podocitos/citología , Transcriptoma
12.
Cell Biosci ; 2(1): 33, 2012 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-22995171

RESUMEN

Nuclear receptors are a family of ligand-activated, DNA sequence-specific transcription factors that regulate various aspects of animal development, cell proliferation, differentiation, and homeostasis. The physiological roles of nuclear receptors and their ligands have been intensively studied in cancer and metabolic syndrome. However, their role in kidney diseases is still evolving, despite their ligands being used clinically to treat renal diseases for decades. This review will discuss the progress of our understanding of the role of nuclear receptors and their ligands in kidney physiology with emphasis on their roles in treating glomerular disorders and podocyte injury repair responses.

13.
J Biol Chem ; 287(42): 35418-35429, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-22908231

RESUMEN

α-Actinins (ACTNs) are a family of proteins cross-linking actin filaments that maintain cytoskeletal organization and cell motility. Recently, it has also become clear that ACTN4 can function in the nucleus. In this report, we found that ACTN4 (full length) and its spliced isoform ACTN4 (Iso) possess an unusual LXXLL nuclear receptor interacting motif. Both ACTN4 (full length) and ACTN4 (Iso) potentiate basal transcription activity and directly interact with estrogen receptor α, although ACTN4 (Iso) binds ERα more strongly. We have also found that both ACTN4 (full length) and ACTN4 (Iso) interact with the ligand-independent and the ligand-dependent activation domains of estrogen receptor α. Although ACTN4 (Iso) interacts efficiently with transcriptional co-activators such as p300/CBP-associated factor (PCAF) and steroid receptor co-activator 1 (SRC-1), the full length ACTN4 protein either does not or does so weakly. More importantly, the flanking sequences of the LXXLL motif are important not only for interacting with nuclear receptors but also for the association with co-activators. Taken together, we have identified a novel extended LXXLL motif that is critical for interactions with both receptors and co-activators. This motif functions more efficiently in a spliced isoform of ACTN4 than it does in the full-length protein.


Asunto(s)
Actinina/metabolismo , Empalme Alternativo/fisiología , Receptor alfa de Estrógeno/metabolismo , Coactivador 1 de Receptor Nuclear/metabolismo , Transcripción Genética/fisiología , Factores de Transcripción p300-CBP/metabolismo , Actinina/genética , Secuencias de Aminoácidos , Línea Celular Tumoral , Receptor alfa de Estrógeno/genética , Humanos , Coactivador 1 de Receptor Nuclear/genética , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Factores de Transcripción p300-CBP/genética
14.
J Biol Chem ; 287(15): 12027-35, 2012 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-22351778

RESUMEN

Mutations in α-actinin 4 (ACTN4) are linked to familial forms of focal segmental glomerulosclerosis (FSGS), a kidney disease characterized by proteinuria due to podocyte injury. The mechanisms underlying ACTN4 mutant-associated FSGS are not completely understood. Although α-actinins are better known to cross-link actin filaments and modulate cytoskeletal organization, we have previously shown that ACTN4 interacts with transcription factors including estrogen receptor and MEF2s and potentiates their transcriptional activity. Nuclear receptors including retinoic acid receptor (RAR) have been proposed to play a protective role in podocytes. We show here that ACTN4 interacts with and enhances transcriptional activation by RARα. In addition, FSGS-linked ACTN4 mutants not only mislocalized to the cytoplasm, but also lost their ability to associate with nuclear receptors. Consequently, FSGS-linked ACTN4 mutants failed to potentiate transcriptional activation by nuclear hormone receptors in podocytes. In addition, overexpression of these mutants suppressed the transcriptional activity mediated by endogenous wild-type ACTN4 possibly by a cytoplasmic sequestration mechanism. Our data provide the first link between FSGS-linked ACTN4 mutants and transcriptional activation by nuclear receptor such as RARα and peroxisome proliferator-activated receptor γ.


Asunto(s)
Actinina/genética , Glomeruloesclerosis Focal y Segmentaria/genética , Mutación Missense , Receptores de Ácido Retinoico/metabolismo , Transcripción Genética , Actinina/metabolismo , Línea Celular , Humanos , Proteínas Mutantes/metabolismo , PPAR gamma/metabolismo , Podocitos/metabolismo , Unión Proteica , Transporte de Proteínas , Receptores de Ácido Retinoico/agonistas , Receptores de Ácido Retinoico/genética , Proteínas Recombinantes/metabolismo , Receptor alfa de Ácido Retinoico , Activación Transcripcional , Tretinoina/fisiología
15.
J Biol Chem ; 286(3): 1850-9, 2011 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-21078666

RESUMEN

Alpha actinins (ACTNs) are known for their ability to modulate cytoskeletal organization and cell motility by cross-linking actin filaments. We show here that ACTN4 harbors a functional LXXLL receptor interaction motif, interacts with nuclear receptors in vitro and in mammalian cells, and potently activates transcription mediated by nuclear receptors. Whereas overexpression of ACTN4 potentiates estrogen receptor α (ERα)-mediated transcription in transient transfection reporter assays, knockdown of ACTN4 decreases it. In contrast, histone deacetylase 7 (HDAC7) inhibits estrogen receptor α (ERα)-mediated transcription. Moreover, the ACTN4 mutant lacking the CaM (calmodulin)-like domain that is required for its interaction with HDAC7 fails to activate transcription by ERα. Chromatin immunoprecipitation (ChIP) assays demonstrate that maximal associations of ACTN4 and HDAC7 with the pS2 promoter are mutually exclusive. Knockdown of ACTN4 significantly decreases the expression of ERα target genes including pS2 and PR and also affects cell proliferation of MCF-7 breast cancer cells with or without hormone, whereas knockdown of HDAC7 exhibits opposite effects. Interestingly, overexpression of wild-type ACTN4, but not the mutants defective in interacting with ERα or HDAC7, results in an increase in pS2 and PR mRNA accumulation in a hormone-dependent manner. In summary, we have identified ACTN4 as a novel, atypical coactivator that regulates transcription networks to control cell growth.


Asunto(s)
Actinina/metabolismo , Neoplasias de la Mama/metabolismo , Proliferación Celular , Receptor alfa de Estrógeno/metabolismo , Histona Desacetilasas/metabolismo , Proteínas de Neoplasias/metabolismo , Actinina/genética , Secuencias de Aminoácidos , Neoplasias de la Mama/genética , Línea Celular Tumoral , Receptor alfa de Estrógeno/genética , Femenino , Técnicas de Silenciamiento del Gen , Histona Desacetilasas/genética , Humanos , Proteínas de Neoplasias/genética , Presenilina-2/biosíntesis , Presenilina-2/genética , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína
16.
J Biol Chem ; 281(46): 35070-80, 2006 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-16980305

RESUMEN

Histone deacetylase 7 (HDAC7) is a member of class IIa HDACs that regulate myocyte enhancer factor-2 (MEF2)-mediated transcription and participate in multiple cellular processes such as T cell apoptosis. We have identified alpha-actinin 1 and 4 as class IIa HDAC-interacting proteins. The interaction domains are mapped to C terminus of alpha-actinin 4 and amino acids 72-172 of HDAC7. A point mutation in HDAC7 that disrupts its association with MEF2A also disrupts its association with alpha-actinin 4, indicating that MEF2A and alpha-actinin 4 binding sites largely overlap. We have also isolated a novel splice variant of alpha-actinin 4 that is predominantly localized in the nucleus, a pattern distinct from the full-length alpha-actinin 4, which is primarily distributed in the cytoplasm and plasma membrane. Using small interfering RNA, chromatin immunoprecipitation, and transient transfection assays, we show that alpha-actinin 4 potentiates expression of TAF55, a putative MEF2 target gene. Loss of MEF2A interaction correlates with loss of the ability of alpha-actinin 4 to potentiate TAF55 promoter activity. Ectopic expression of alpha-actinin 4, but not the mutant defective in MEF2A association, leads to disruption of HDAC7.MEF2A association and enhancement of MEF2-mediated transcription. Taken together, we have identified a novel mechanism by which HDAC7 activity is negatively regulated and uncovered a previously unknown function of alpha-actinin 4.


Asunto(s)
Actinina/metabolismo , Histona Desacetilasas/metabolismo , Proteínas de Dominio MADS/metabolismo , Proteínas de Microfilamentos/metabolismo , Factores Reguladores Miogénicos/metabolismo , Secuencia de Aminoácidos , Células HeLa , Humanos , Proteínas de Dominio MADS/genética , Factores de Transcripción MEF2 , Factores Reguladores Miogénicos/genética , Transcripción Genética , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA