Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Nucleic Acids Res ; 51(13): 6509-6527, 2023 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-36940725

RESUMEN

Telomere maintenance is a hallmark of malignant cells and allows cancers to divide indefinitely. In some cancers, this is achieved through the alternative lengthening of telomeres (ALT) pathway. Whilst loss of ATRX is a near universal feature of ALT-cancers, it is insufficient in isolation. As such, other cellular events must be necessary - but the exact nature of the secondary events has remained elusive. Here, we report that trapping of proteins (such as TOP1, TOP2A and PARP1) on DNA leads to ALT induction in cells lacking ATRX. We demonstrate that protein-trapping chemotherapeutic agents, such as etoposide, camptothecin and talazoparib, induce ALT markers specifically in ATRX-null cells. Further, we show that treatment with G4-stabilising drugs cause an increase in trapped TOP2A levels which leads to ALT induction in ATRX-null cells. This process is MUS81-endonuclease and break-induced replication dependent, suggesting that protein trapping leads to replication fork stalling, with these forks being aberrantly processed in the absence of ATRX. Finally, we show ALT-positive cells harbour a higher load of genome-wide trapped proteins, such as TOP1, and knockdown of TOP1 reduced ALT activity. Taken together, these findings suggest that protein trapping is a fundamental driving force behind ALT-biology in ATRX-deficient malignancies.


A key feature of all cancer cells is their ability to divide indefinitely, and this is dependent on circumvention of telomere shortening through induction of a telomere maintenance mechanism, such as the telomerase-independent, Alternative Lengthening of Telomeres (ALT) pathway. The ALT pathway is characterised by loss of the ATRX chromatin remodeler. The current study provides evidence that, in the absence of ATRX, increased trapping of proteins on DNA leads to replication fork stalling and collapse. At telomeres, this leads to ALT pathway activity. These results help to better understand ALT tumours and might, eventually, be instrumental in developing new therapeutic strategies.


Asunto(s)
Neoplasias , Telómero , Humanos , ADN , Neoplasias/genética , Telomerasa/genética , Telómero/genética , Telómero/metabolismo , Homeostasis del Telómero , Proteína Nuclear Ligada al Cromosoma X/genética , Proteína Nuclear Ligada al Cromosoma X/metabolismo
2.
Mol Cell ; 55(5): 733-44, 2014 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-25127513

RESUMEN

Gene transcription responds to stress and metabolic signals to optimize growth and survival. Histone H3 (H3) lysine 4 trimethylation (K4me3) facilitates state changes, but how levels are coordinated with the environment is unclear. Here, we show that isomerization of H3 at the alanine 15-proline 16 (A15-P16) peptide bond is influenced by lysine 14 (K14) and controls gene-specific K4me3 by balancing the actions of Jhd2, the K4me3 demethylase, and Spp1, a subunit of the Set1 K4 methyltransferase complex. Acetylation at K14 favors the A15-P16trans conformation and reduces K4me3. Environmental stress-induced genes are most sensitive to the changes at K14 influencing H3 tail conformation and K4me3. By contrast, ribosomal protein genes maintain K4me3, required for their repression during stress, independently of Spp1, K14, and P16. Thus, the plasticity in control of K4me3, via signaling to K14 and isomerization at P16, informs distinct gene regulatory mechanisms and processes involving K4me3.


Asunto(s)
Lisina/metabolismo , Prolina/metabolismo , Saccharomyces cerevisiae/genética , Cromatina/química , Cromatina/metabolismo , Epigénesis Genética , Histonas/química , Histonas/metabolismo , Isomerismo , Lisina/química , Prolina/química , Estructura Terciaria de Proteína , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Estrés Fisiológico
3.
EMBO Rep ; 18(6): 914-928, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28487353

RESUMEN

ATRX is a chromatin remodelling factor found at a wide range of tandemly repeated sequences including telomeres (TTAGGG)n ATRX mutations are found in nearly all tumours that maintain their telomeres via the alternative lengthening of telomere (ALT) pathway, and ATRX is known to suppress this pathway. Here, we show that recruitment of ATRX to telomeric repeats depends on repeat number, orientation and, critically, on repeat transcription. Importantly, the transcribed telomeric repeats form RNA-DNA hybrids (R-loops) whose abundance correlates with the recruitment of ATRX Here, we show loss of ATRX is also associated with increased R-loop formation. Our data suggest that the presence of ATRX at telomeres may have a central role in suppressing deleterious DNA secondary structures that form at transcribed telomeric repeats, and this may account for the increased DNA damage, stalling of replication and homology-directed repair previously observed upon loss of ATRX function.


Asunto(s)
Ensamble y Desensamble de Cromatina , ADN/genética , ARN/genética , Telómero/genética , Telómero/metabolismo , Proteína Nuclear Ligada al Cromosoma X/metabolismo , Cromatina , ADN/química , Daño del ADN , Replicación del ADN , G-Cuádruplex , Humanos , Homeostasis del Telómero/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Proteína Nuclear Ligada al Cromosoma X/deficiencia , Proteína Nuclear Ligada al Cromosoma X/genética
4.
Trends Biochem Sci ; 38(9): 461-6, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23916100

RESUMEN

The regulation of chromatin structure is of paramount importance for a variety of fundamental nuclear processes, including gene expression, DNA repair, replication, and recombination. The ATP-dependent chromatin-remodelling factor ATRX (α thalassaemia/mental retardation X-linked) has emerged as a key player in each of these processes. Exciting recent developments suggest that ATRX plays a variety of key roles at tandem repeat sequences within the genome, including the deposition of a histone variant, prevention of replication fork stalling, and the suppression of a homologous recombination-based pathway of telomere maintenance. Here, we provide a mechanistic overview of the role of ATRX in each of these processes, and propose how they may be connected to give rise to seemingly disparate human diseases.


Asunto(s)
Cromatina/metabolismo , ADN Helicasas/metabolismo , Proteínas Nucleares/metabolismo , Ensamble y Desensamble de Cromatina , ADN Helicasas/genética , Replicación del ADN/genética , Replicación del ADN/fisiología , Histonas/metabolismo , Humanos , Proteínas Nucleares/genética , Telómero/metabolismo , Proteína Nuclear Ligada al Cromosoma X
5.
Proc Natl Acad Sci U S A ; 111(42): 15190-5, 2014 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-25288776

RESUMEN

Cancers result from the accumulation of genetic lesions, but the cellular consequences of driver mutations remain unclear, especially during the earliest stages of malignancy. The V617F mutation in the JAK2 non-receptor tyrosine kinase (JAK2V617F) is present as an early somatic event in most patients with myeloproliferative neoplasms (MPNs), and the study of these chronic myeloid malignancies provides an experimentally tractable approach to understanding early tumorigenesis. Introduction of exogenous JAK2V617F impairs replication fork progression and is associated with activation of the intra-S checkpoint, with both effects mediated by phosphatidylinositide 3-kinase (PI3K) signaling. Analysis of clonally derived JAK2V617F-positive erythroblasts from MPN patients also demonstrated impaired replication fork progression accompanied by increased levels of replication protein A (RPA)-containing foci. However, the associated intra-S checkpoint response was impaired in erythroblasts from polycythemia vera (PV) patients, but not in those from essential thrombocythemia (ET) patients. Moreover, inhibition of p53 in PV erythroblasts resulted in more gamma-H2Ax (γ-H2Ax)-marked double-stranded breaks compared with in like-treated ET erythroblasts, suggesting the defective intra-S checkpoint function seen in PV increases DNA damage in the context of attenuated p53 signaling. These results demonstrate oncogene-induced impairment of replication fork progression in primary cells from MPN patients, reveal unexpected disease-restricted differences in activation of the intra-S checkpoint, and have potential implications for the clonal evolution of malignancies.


Asunto(s)
Puntos de Control del Ciclo Celular , Replicación del ADN , Janus Quinasa 2/fisiología , Fase S , Apoptosis , División Celular , Cromosomas/metabolismo , Cromosomas/ultraestructura , Daño del ADN , Reparación del ADN , Diploidia , Fibroblastos/metabolismo , Genotipo , Enfermedades Hematológicas/genética , Humanos , Janus Quinasa 2/genética , Leucemia/metabolismo , Leucemia/patología , Microscopía Fluorescente , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , Fosforilación , ARN Interferente Pequeño/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
6.
Front Cell Dev Biol ; 11: 1175069, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37342232

RESUMEN

Hereditary fibrosing poikiloderma (HFP) is a rare human dominant negative disorder caused by mutations in the FAM111B gene that encodes a nuclear trypsin-like serine protease. HFP patients present with symptoms including skin abnormalities, tendon contractures, myopathy and lung fibrosis. We characterized the cellular roles of human FAM111B using U2OS and MCF7 cell lines and report here that the protease interacts with components of the nuclear pore complex. Loss of FAM111B expression resulted in abnormal nuclear shape and reduced telomeric DNA content suggesting that FAM111B protease is required for normal telomere length; we show that this function is independent of telomerase or recombination driven telomere extension. Even though FAM111B-deficient cells were proficient in DNA repair, they showed hallmarks of genomic instability such as increased levels of micronuclei and ultra-fine DNA bridges. When mutated as in HFP, FAM111B was more frequently localized to the nuclear envelope, suggesting that accumulation of the mutated protease at the nuclear periphery may drive the disease pathology.

7.
Curr Opin Genet Dev ; 18(2): 116-22, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18295475

RESUMEN

Post-translational modifications to histone proteins and methylation of DNA comprise the epigenome of a cell. The epigenome, which changes through development, controls access to our genes. Recent advances in DNA sequencing technology has led to genome-wide distribution data for a limited number of histone modifications in mammalian stem cells and some differentiated lineages. These studies reveal predictive correlations between histone modifications, different classes of gene and chromosomal features. Moreover, this glimpse into our epigenome challenges current ideas about regulation of gene expression. Many genes in stem cells are poised for expression with initiated RNA polymerase II at the promoter. This state is maintained by an epigenetic mark through multiple lineages until the gene is expressed.


Asunto(s)
Epigénesis Genética/genética , Animales , Diferenciación Celular , Histonas/metabolismo , Humanos , Metilación , ARN Polimerasa II/metabolismo , Transcripción Genética/genética
8.
Genes (Basel) ; 12(11)2021 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-34828344

RESUMEN

The study of the molecular pathways underlying cancer has given us important insights into how breaks in our DNA are repaired and the dire consequences that can occur when these processes are perturbed. Extensive research over the past 20 years has shown that the key molecular event underpinning a subset of cancers involves the deregulated repair of DNA double-strand breaks (DSBs) at telomeres, which in turn leads to telomere lengthening and the potential for replicative immortality. Here we discuss, in-depth, recent major breakthroughs in our understanding of the mechanisms underpinning this pathway known as the alternative lengthening of telomeres (ALT). We explore how this gives us important insights into how DSB repair at telomeres is regulated, with relevance to the cell-cycle-dependent regulation of repair, repair of stalled replication forks and the spatial regulation of DSB repair.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Replicación del ADN , Homeostasis del Telómero , Telómero/metabolismo , Ciclo Celular , ADN/metabolismo
9.
Front Oncol ; 9: 1518, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32039009

RESUMEN

Achieving replicative immortality is a crucial step in tumorigenesis and requires both bypassing cell cycle checkpoints and the extension of telomeres, sequences that protect the distal ends of chromosomes during replication. In the majority of cancers this is achieved through the enzyme telomerase, however a subset of cancers instead utilize a telomerase-independent mechanism of telomere elongation-the Alternative Lengthening of Telomeres (ALT) pathway. Recent work has aimed to decipher the exact mechanism that underlies this pathway. To this end, this pathway has now been shown to extend telomeres through exploitation of DNA repair machinery in a unique process that may present a number of druggable targets. The identification of such targets, and the subsequent development or repurposing of therapies to these targets may be crucial to improving the prognosis for many ALT-positive cancers, wherein mean survival is lower than non-ALT counterparts and the cancers themselves are particularly unresponsive to standard of care therapies. In this review we summarize the recent identification of many aspects of the ALT pathway, and the therapies that may be employed to exploit these new targets.

10.
Nat Commun ; 6: 7538, 2015 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-26143912

RESUMEN

Fifteen per cent of cancers maintain telomere length independently of telomerase by the homologous recombination (HR)-associated alternative lengthening of telomeres (ALT) pathway. A unifying feature of these tumours are mutations in ATRX. Here we show that expression of ectopic ATRX triggers a suppression of the pathway and telomere shortening. Importantly ATRX-mediated ALT suppression is dependent on the histone chaperone DAXX. Re-expression of ATRX is associated with a reduction in replication fork stalling, a known trigger for HR and loss of MRN from telomeres. A G-quadruplex stabilizer partially reverses the effect of ATRX, inferring ATRX may normally facilitate replication through these sequences that, if they persist, promote ALT. We propose that defective telomere chromatinization through loss of ATRX promotes the persistence of aberrant DNA secondary structures, which in turn present a barrier to DNA replication, leading to replication fork stalling, collapse, HR and subsequent recombination-mediated telomere synthesis in ALT cancers.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , ADN Helicasas/metabolismo , Proteínas Nucleares/metabolismo , Homeostasis del Telómero/fisiología , Línea Celular Tumoral , Células , ADN Helicasas/genética , Replicación del ADN , Humanos , Proteínas Nucleares/genética , Telómero/metabolismo , Proteína Nuclear Ligada al Cromosoma X
11.
PLoS One ; 9(3): e92915, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24651726

RESUMEN

The chromatin remodeling protein ATRX, which targets tandem repetitive DNA, has been shown to be required for expression of the alpha globin genes, for proliferation of a variety of cellular progenitors, for chromosome congression and for the maintenance of telomeres. Mutations in ATRX have recently been identified in tumours which maintain their telomeres by a telomerase independent pathway involving homologous recombination thought to be triggered by DNA damage. It is as yet unknown whether there is a central underlying mechanism associated with ATRX dysfunction which can explain the numerous cellular phenomena observed. There is, however, growing evidence for its role in the replication of various repetitive DNA templates which are thought to have a propensity to form secondary structures. Using a mouse knockout model we demonstrate that ATRX plays a direct role in facilitating DNA replication. Ablation of ATRX alone, although leading to a DNA damage response at telomeres, is not sufficient to trigger the alternative lengthening of telomere pathway in mouse embryonic stem cells.


Asunto(s)
ADN Helicasas/metabolismo , Replicación del ADN , Proteínas Nucleares/metabolismo , Animales , Línea Celular , Ensamble y Desensamble de Cromatina , Roturas del ADN de Doble Cadena , Daño del ADN , ADN Helicasas/genética , Células Madre Embrionarias/metabolismo , Técnicas de Inactivación de Genes , Humanos , Ratones , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/genética , Fase S , Telómero/metabolismo , Proteína Nuclear Ligada al Cromosoma X
12.
Curr Opin Genet Dev ; 23(3): 289-94, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23453691

RESUMEN

Understanding the underlying molecular basis for disease can often be a prolonged and tortuous process with many false leads and blind alleys. Relating the cause of ATR-X syndrome to the function of the protein ATRX is a case in point. In this review we attempt to bring together the diverse biological phenomena associated with ATRX dysfunction with what has recently been discovered concerning the chromatin remodelling activity of this protein. This potentially casts light on how defective DNA replication/histone replacement can impact on transcription, telomere maintenance and also possibly chromosome segregation.


Asunto(s)
ADN Helicasas/genética , Replicación del ADN/genética , Discapacidad Intelectual Ligada al Cromosoma X/genética , Proteínas Nucleares/genética , Homeostasis del Telómero/genética , Talasemia alfa/genética , Ensamble y Desensamble de Cromatina/genética , Segregación Cromosómica , Histonas/genética , Histonas/metabolismo , Humanos , Discapacidad Intelectual Ligada al Cromosoma X/etiología , Discapacidad Intelectual Ligada al Cromosoma X/patología , Mutación , Transcripción Genética , Proteína Nuclear Ligada al Cromosoma X , Talasemia alfa/etiología , Talasemia alfa/patología
13.
Nat Struct Mol Biol ; 18(7): 777-82, 2011 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-21666677

RESUMEN

Accurate read-out of chromatin modifications is essential for eukaryotic life. Mutations in the gene encoding X-linked ATRX protein cause a mental-retardation syndrome, whereas wild-type ATRX protein targets pericentric and telomeric heterochromatin for deposition of the histone variant H3.3 by means of a largely unknown mechanism. Here we show that the ADD domain of ATRX, in which most syndrome-causing mutations occur, engages the N-terminal tail of histone H3 through two rigidly oriented binding pockets, one for unmodified Lys4 and the other for di- or trimethylated Lys9. In vivo experiments show this combinatorial readout is required for ATRX localization, with recruitment enhanced by a third interaction through heterochromatin protein-1 (HP1) that also recognizes trimethylated Lys9. The cooperation of ATRX ADD domain and HP1 in chromatin recruitment results in a tripartite interaction that may span neighboring nucleosomes and illustrates how the 'histone-code' is interpreted by a combination of multivalent effector-chromatin interactions.


Asunto(s)
ADN Helicasas/química , Heterocromatina/metabolismo , Histonas/metabolismo , Proteínas Nucleares/química , Sitios de Unión , Ensamble y Desensamble de Cromatina , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , ADN Helicasas/metabolismo , ADN Helicasas/fisiología , Heterocromatina/química , Código de Histonas , Histonas/química , Metilación , Resonancia Magnética Nuclear Biomolecular , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiología , Proteína Nuclear Ligada al Cromosoma X
14.
Mol Cell Biol ; 29(9): 2419-30, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19273607

RESUMEN

The SANT domain is a nucleosome recognition module found in transcriptional regulatory proteins, including chromatin-modifying enzymes. It shows high functional degeneracy between species, varying in sequence and copy number. Here, we investigate functions in vivo associated with two SANT motifs, SANT and SLIDE, in the Saccharomyces cerevisiae Isw1 chromatin-remodeling ATPase. We show that differences in the primary structures of the SANT and SLIDE domains in yeast and Drosophila melanogaster reflect their different functions. In yeast, the SLIDE domain is required for histone interactions, while this is a function of the SANT domain in flies. In yeast, both motifs are required for optimal association with chromatin and for formation of the Isw1b complex (Isw1, Ioc2, and Ioc4). Moreover, nucleosome remodeling at the MET16 locus is defective in strains lacking the SANT or SLIDE domain. In contrast, the SANT domain is dispensable for the interaction between Isw1 and Ioc3 in the Isw1a complex. We show that, although defective in nucleosome remodeling, Isw1 lacking the SANT domain is able to repress transcription initiation at the MET16 promoter. Thus, chromatin remodeling and transcriptional repression are distinct activities of Isw1.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Ensamble y Desensamble de Cromatina , Proteínas de Unión al ADN/metabolismo , Regulación Fúngica de la Expresión Génica , Nucleosomas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae , Adenosina Trifosfatasas/genética , Secuencia de Aminoácidos , Animales , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Histonas/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Estructura Terciaria de Proteína , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Alineación de Secuencia
15.
Mol Cell Biol ; 28(8): 2840-9, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18268010

RESUMEN

Histone modifications occur in precise patterns and are proposed to signal the recruitment of effector molecules that profoundly impact chromatin structure, gene regulation, and cell cycle events. The linked modifications serine 10 phosphorylation and lysine 14 acetylation on histone H3 (H3S10phK14ac), modifications conserved from Saccharomyces cerevisiae to humans, are crucial for transcriptional activation of many genes. However, the mechanism of H3S10phK14ac involvement in these processes is unclear. To shed light on the role of this dual modification, we utilized H3 peptide affinity assays to identify H3S10phK14ac-interacting proteins. We found that the interaction of the known phospho-binding 14-3-3 proteins with H3 is dependent on the presence of both of these marks, not just phosphorylation alone. This is true of mammalian 14-3-3 proteins as well as the yeast homologues Bmh1 and Bmh2. The importance of acetylation in this interaction is also seen in vivo, where K14 acetylation is required for optimal Bmh1 recruitment to the GAL1 promoter during transcriptional activation.


Asunto(s)
Proteínas 14-3-3/metabolismo , Histonas/metabolismo , Acetilación , Calorimetría , Células HeLa , Humanos , Fosforilación , Fosfoserina/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
16.
Mol Cell ; 20(2): 199-211, 2005 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-16246723

RESUMEN

Phosphorylation of histone H3 is implicated in transcriptional activation and chromosome condensation, but its immediate molecular function has remained obscure. By affinity chromatography of nuclear extracts against modified H3 tail peptides, we identified 14-3-3 isoforms as proteins that bind these tails in a strictly phosphorylation-dependent manner. Acetylation of lysines 9 and 14 does not impede 14-3-3 binding to serine 10-phosphorylated H3 tails. In vivo, 14-3-3 is inducibly recruited to c-fos and c-jun nucleosomes upon gene activation, concomitant with H3 phosphoacetylation. We have determined the structures of 14-3-3zeta complexed with serine 10-phosphorylated or phosphoacetylated H3 peptides. These reveal a distinct mode of 14-3-3/phosphopeptide binding and provide a structural understanding for the lack of effect of acetylation at lysines 9 and 14 on this interaction. 14-3-3 isoforms thus represent a class of proteins that mediate the effect of histone phosphorylation at inducible genes.


Asunto(s)
Proteínas 14-3-3/metabolismo , Histonas/metabolismo , Proteínas 14-3-3/química , Acetilación , Secuencia de Aminoácidos , Animales , Línea Celular , Cristalografía por Rayos X , Células HeLa , Histonas/química , Histonas/aislamiento & purificación , Humanos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Nucleosomas/genética , Nucleosomas/metabolismo , Fosforilación , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Alineación de Secuencia , Serina/química , Serina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA