Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Hum Mol Genet ; 32(23): 3225-3236, 2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-37642363

RESUMEN

Oligodendrocytes (OLs) produce myelin sheaths around axons in the central nervous system (CNS). Myelin accelerates the propagation of action potentials along axons and supports the integrity of axons. Impaired myelination has been linked to neurological and neuropsychiatric disorders. As a major component of CNS myelin, 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNP) plays an indispensable role in the axon-supportive function of myelin. Notably, this function requires a high-level expression of CNP in OLs, as evidenced by downregulated expression of CNP in mental disorders and animal models. Little is known about how CNP expression is regulated in OLs. Especially, OL enhancers that govern CNP remain elusive. We have recently developed a powerful method that links OL enhancers to target genes in a principled manner. Here, we applied it to Cnp, uncovering two OL enhancers for it (termed Cnp-E1 and Cnp-E2). Epigenome editing analysis revealed that Cnp-E1 and Cnp-E2 are dedicated to Cnp. ATAC-seq and ChIP-seq data show that Cnp-E1 and Cnp-E2 are conserved OL-specific enhancers. Single cell multi-omics data that jointly profile gene expression and chromatin accessibility suggest that Cnp-E2 plays an important role in Cnp expression in the early stage of OL differentiation while Cnp-E1 sustains it in mature OLs.


Asunto(s)
Vaina de Mielina , Oligodendroglía , Animales , Humanos , Oligodendroglía/metabolismo , Vaina de Mielina/genética , Vaina de Mielina/metabolismo , Sistema Nervioso Central , Axones/fisiología , Diferenciación Celular/genética
2.
Hum Mol Genet ; 32(5): 835-846, 2023 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-36193754

RESUMEN

Olig2 is a basic helix-loop-helix transcription factor that plays a critical role in the central nervous system. It directs the specification of motor neurons and oligodendrocyte precursor cells (OPCs) from neural progenitors and the subsequent maturation of OPCs into myelin-forming oligodendrocytes (OLs). It is also required for the development of astrocytes. Despite a decade-long search, enhancers that regulate the expression of Olig2 remain elusive. We have recently developed an innovative method that maps promoter-distal enhancers to genes in a principled manner. Here, we applied it to Olig2 in the context of OL lineage cells, uncovering an OL enhancer for it (termed Olig2-E1). Silencing Olig2-E1 by CRISPRi epigenome editing significantly downregulated Olig2 expression. Luciferase assay and ATAC-seq and ChIP-seq data show that Olig2-E1 is an OL-specific enhancer that is conserved across human, mouse and rat. Hi-C data reveal that Olig2-E1 physically interacts with OLIG2 and suggest that this interaction is specific to OL lineage cells. In sum, Olig2-E1 is an evolutionarily conserved OL-specific enhancer that drives the expression of Olig2.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Proteínas del Tejido Nervioso , Ratones , Ratas , Animales , Humanos , Proteínas del Tejido Nervioso/genética , Diferenciación Celular/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Oligodendroglía/metabolismo , Vaina de Mielina/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo
3.
Hum Mol Genet ; 30(23): 2225-2239, 2021 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-34230963

RESUMEN

Oligodendrocytes (OLs) produce myelin in the central nervous system (CNS), which accelerates the propagation of action potentials and supports axonal integrity. As a major component of CNS myelin, proteolipid protein 1 (Plp1) is indispensable for the axon-supportive function of myelin. Notably, this function requires the continuous high-level expression of Plp1 in OLs. Equally important is the controlled expression of Plp1, as illustrated by Pelizaeus-Merzbacher disease for which the most common cause is PLP1 overexpression. Despite a decade-long search, promoter-distal OL enhancers that govern Plp1 remain elusive. We have recently developed an innovative method that maps promoter-distal enhancers to genes in a principled manner. Here, we applied it to Plp1, uncovering two OL enhancers for it (termed Plp1-E1 and Plp1-E2). Remarkably, clustered regularly interspaced short palindromic repeats (CRISPR) interference epigenome editing showed that Plp1-E1 and Plp1-E2 do not regulate two genes in their vicinity, highlighting their exquisite specificity to Plp1. Assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) and chromatin immunoprecipitation with high-throughput sequencing (ChIP-seq) data show that Plp1-E1 and Plp1-E2 are OL-specific enhancers that are conserved among human, mouse and rat. Hi-C data reveal that the physical interactions between Plp1-E1/2 and PLP1 are among the strongest in OLs and specific to OLs. We also show that Myrf, a master regulator of OL development, acts on Plp1-E1 and Plp1-E2 to promote Plp1 expression.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Proteína Proteolipídica de la Mielina/genética , Oligodendroglía/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Sistemas CRISPR-Cas , Secuenciación de Inmunoprecipitación de Cromatina , Mapeo Cromosómico , Proteínas de Unión al ADN/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Proteína Proteolipídica de la Mielina/metabolismo , Motivos de Nucleótidos , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo
4.
J Biol Chem ; 296: 100612, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33798553

RESUMEN

Myrf is a pleiotropic membrane-bound transcription factor that plays critical roles in diverse organisms, including in oligodendrocyte differentiation, embryonic development, molting, and synaptic plasticity. Upon autolytic cleavage, the Myrf N-terminal fragment enters the nucleus as a homo-trimer and functions as a transcription factor. Homo-trimerization is essential for this function because it imparts DNA-binding specificity and affinity. Recent exome sequencing studies have implicated four de novo MYRF DNA-binding domain (DBD) mutations (F387S, Q403H, G435R, and L479V) in novel syndromic birth defects involving the diaphragm, heart, and the urogenital tract. It remains unknown whether and how these four mutations alter the transcription factor function of MYRF. Here, we studied them by introducing homologous mutations to the mouse Myrf protein. We found that the four DBD mutations abolish the transcriptional activity of the Myrf N-terminal fragment by interfering with its homo-trimerization ability by perturbing the DBD structure. Since the Myrf N-terminal fragment strictly functions as a homo-trimer, any loss-of-function mutation has the potential to act as a dominant negative. We observed that one copy of Myrf-F387S, Myrf-Q403H, or Myrf-L479V, but not Myrf-G435R, was tolerated by the Myrf N-terminal homo-trimer for structural and functional integrity. These data suggest that F387S, Q403H, and L479V cause birth defects by haploinsufficiency, while G435R does so via dominant negative functionality.


Asunto(s)
Anomalías Congénitas/genética , Proteínas de la Membrana/metabolismo , Mutación , Oligodendroglía/metabolismo , Factores de Transcripción/metabolismo , Animales , Células Cultivadas , Anomalías Congénitas/patología , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Oligodendroglía/citología , Conformación Proteica , Dominios Proteicos , Factores de Transcripción/química , Factores de Transcripción/genética
5.
Hum Mol Genet ; 28(21): 3600-3609, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31509193

RESUMEN

Multiple sclerosis (MS) is known as an autoimmune demyelinating disease of the central nervous system. However, its cause remains elusive. Given previous studies suggesting that dysfunctional oligodendrocytes (OLs) may trigger MS, we tested whether single nucleotide polymorphisms (SNPs) associated with MS affect OL enhancers, potentially increasing MS risk by dysregulating gene expression of OL lineage cells. We found that two closely spaced OL enhancers, which are 3 Kb apart on chromosome 13, overlap two MS SNPs in linkage disequilibrium-rs17594362 and rs12429256. Our data revealed that the two MS SNPs significantly up-regulate the associated OL enhancers, which we have named as Rgcc-E1 and Rgcc-E2. Analysis of Hi-C data and epigenome editing experiments shows that Rgcc is the primary target of Rgcc-E1 and Rgcc-E2. Collectively, these data indicate that the molecular mechanism of rs17594362 and rs12429256 is to induce Rgcc overexpression by potentiating the enhancer activity of Rgcc-E1 and Rgcc-E2. Importantly, the dosage of the rs17594362/rs12429256 risk allele is positively correlated with the expression level of Rgcc in the human population, confirming our molecular mechanism. Our study also suggests that Rgcc overexpression in OL lineage cells may be a key cellular mechanism of rs17594362 and rs12429256 for MS.


Asunto(s)
Esclerosis Múltiple/genética , Polimorfismo de Nucleótido Simple , Alelos , Cromosomas Humanos Par 13/genética , Elementos de Facilitación Genéticos , Predisposición Genética a la Enfermedad , Humanos , Desequilibrio de Ligamiento
6.
Nucleic Acids Res ; 45(9): 5112-5125, 2017 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-28160598

RESUMEN

Myrf is a key transcription factor for oligodendrocyte differentiation and central nervous system myelination. We and others have previously shown that Myrf is generated as a membrane protein in the endoplasmic reticulum (ER), and that it undergoes auto-processing to release its N-terminal fragment from the ER, which enters the nucleus to work as a transcription factor. These previous studies allow a glimpse into the unusual complexity behind the biogenesis and function of the transcription factor domain of Myrf. Here, we report that Myrf N-terminal fragments assemble into stable homo-trimers before ER release. Consequently, Myrf N-terminal fragments are released from the ER only as homo-trimers. Our re-analysis of a previous genetic screening result in Caenorhabditis elegans shows that homo-trimerization is essential for the biological functions of Myrf N-terminal fragment, and that the region adjacent to the DNA-binding domain is pivotal to its homo-trimerization. Further, our computational analysis uncovered a novel homo-trimeric DNA motif that mediates the homo-trimeric DNA binding of Myrf N-terminal fragments. Importantly, we found that homo-trimerization defines the DNA binding specificity of Myrf N-terminal fragments. In sum, our study elucidates the molecular mechanism governing the biogenesis and function of Myrf N-terminal fragments and its physiological significance.


Asunto(s)
Diferenciación Celular , Proteínas de la Membrana/fisiología , Oligodendroglía/citología , Multimerización de Proteína , Factores de Transcripción/fisiología , Animales , Línea Celular , ADN/metabolismo , Retículo Endoplásmico/metabolismo , Células HEK293 , Células HeLa , Humanos , Ratones , Oligodendroglía/metabolismo , Fragmentos de Péptidos/química , Unión Proteica , Dominios Proteicos , Ratas , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Transcripción Genética
7.
PLoS Biol ; 11(8): e1001624, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23966832

RESUMEN

Myelination of the central nervous system (CNS) is critical to vertebrate nervous systems for efficient neural signaling. CNS myelination occurs as oligodendrocytes terminally differentiate, a process regulated in part by the myelin regulatory factor, MYRF. Using bioinformatics and extensive biochemical and functional assays, we find that MYRF is generated as an integral membrane protein that must be processed to release its transcription factor domain from the membrane. In contrast to most membrane-bound transcription factors, MYRF proteolysis seems constitutive and independent of cell- and tissue-type, as we demonstrate by reconstitution in E. coli and yeast. The apparent absence of physiological cues raises the question as to how and why MYRF is processed. By using computational methods capable of recognizing extremely divergent sequence homology, we identified a MYRF protein domain distantly related to bacteriophage tailspike proteins. Although occurring in otherwise unrelated proteins, the phage domains are known to chaperone the tailspike proteins' trimerization and auto-cleavage, raising the hypothesis that the MYRF domain might contribute to a novel activation method for a membrane-bound transcription factor. We find that the MYRF domain indeed serves as an intramolecular chaperone that facilitates MYRF trimerization and proteolysis. Functional assays confirm that the chaperone domain-mediated auto-proteolysis is essential both for MYRF's transcriptional activity and its ability to promote oligodendrocyte maturation. This work thus reveals a previously unknown key step in CNS myelination. These data also reconcile conflicting observations of this protein family, different members of which have been identified as transmembrane or nuclear proteins. Finally, our data illustrate a remarkable evolutionary repurposing between bacteriophages and eukaryotes, with a chaperone domain capable of catalyzing trimerization-dependent auto-proteolysis in two entirely distinct protein and cellular contexts, in one case participating in bacteriophage tailspike maturation and in the other activating a key transcription factor for CNS myelination.


Asunto(s)
Proteínas de la Membrana/metabolismo , Factores de Transcripción/metabolismo , Sistema Nervioso Central/metabolismo , Escherichia coli/metabolismo , Humanos , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Unión Proteica , Estructura Terciaria de Proteína
8.
bioRxiv ; 2023 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-37609196

RESUMEN

The role of non-coding regulatory elements and how they might contribute to tissue type specificity of disease phenotypes is poorly understood. Autosomal Dominant Leukodystrophy (ADLD) is a fatal, adult-onset, neurological disorder that is characterized by extensive CNS demyelination. Most cases of ADLD are caused by tandem genomic duplications involving the lamin B1 gene ( LMNB1 ) while a small subset are caused by genomic deletions upstream of the gene. Utilizing data from recently identified families that carry LMNB1 gene duplications but do not exhibit demyelination, ADLD patient tissues, CRISPR modified cell lines and mouse models, we have identified a novel silencer element that is lost in ADLD patients and that specifically targets overexpression to oligodendrocytes. This element consists of CTCF binding sites that mediate three-dimensional chromatin looping involving the LMNB1 and the recruitment of the PRC2 repressor complex. Loss of the silencer element in ADLD identifies a previously unknown role for silencer elements in tissue specificity and disease causation.

9.
Bioinformatics ; 27(21): 3024-8, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21908540

RESUMEN

MOTIVATION: A number of computational methods have been proposed that predict protein-protein interactions (PPIs) based on protein sequence features. Since the number of potential non-interacting protein pairs (negative PPIs) is very high both in absolute terms and in comparison to that of interacting protein pairs (positive PPIs), computational prediction methods rely upon subsets of negative PPIs for training and validation. Hence, the need arises for subset sampling for negative PPIs. RESULTS: We clarify that there are two fundamentally different types of subset sampling for negative PPIs. One is subset sampling for cross-validated testing, where one desires unbiased subsets so that predictive performance estimated with them can be safely assumed to generalize to the population level. The other is subset sampling for training, where one desires the subsets that best train predictive algorithms, even if these subsets are biased. We show that confusion between these two fundamentally different types of subset sampling led one study recently published in Bioinformatics to the erroneous conclusion that predictive algorithms based on protein sequence features are hardly better than random in predicting PPIs. Rather, both protein sequence features and the 'hubbiness' of interacting proteins contribute to effective prediction of PPIs. We provide guidance for appropriate use of random versus balanced sampling. AVAILABILITY: The datasets used for this study are available at http://www.marcottelab.org/PPINegativeDataSampling. CONTACT: yungki@mail.utexas.edu; marcotte@icmb.utexas.edu. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Algoritmos , Mapeo de Interacción de Proteínas/métodos , Biología Computacional/métodos , Humanos , Análisis de Secuencia de Proteína
11.
PLoS One ; 15(7): e0235857, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32730263

RESUMEN

OBJECTIVE: The risk loci for juvenile idiopathic arthritis (JIA) consist of extended haplotypes that include functional elements in addition to canonical coding genes. As with most autoimmune diseases, the risk haplotypes for JIA are highly enriched for H3K4me1/H3K27ac histone marks, epigenetic signatures that typically identify poised or active enhancers. In this study, we test the hypothesis that genetic risk for JIA is exerted through altered enhancer-mediated gene regulation. METHODS: We mined publically available HiC and other chromatin conformation data to determine whether H3K27ac-marked regions in 25 JIA risk loci showed physical evidence of contact with gene promoters. We also used in vitro reporter assays to establish as proof-of-concept the idea that genetic variants in linkage disequilibrium with GWAS-identified tag SNPs alter enhancer function. RESULTS: All 25 loci examined showed multiple contact sites in the 4 different cell lines that we queried. These regions were characterized by HiC-defined loop structures that included 237 immune-related genes. Using in vitro assays, we found that a 657 bp, H3K4me1/H3K27-marked region within the first intron of IL2RA shows enhancer activity in reporter assays, and this activity is attenuated by SNPs on the IL2RA haplotype that we identified using whole genome sequencing of children with JIA. Similarly, we identified a 1,669 bp sequence in an intergenic region of the IL6R locus where SNPs identified in children with JIA increase enhancer function in reporter assays. CONCLUSIONS: These studies provide evidence that altered enhancer function contributes to genetic risk in JIA. Further studies to identify the specific target genes of genetically altered enhancers are warranted.


Asunto(s)
Artritis Juvenil/genética , Ensamble y Desensamble de Cromatina , Elementos de Facilitación Genéticos , Histonas/genética , Polimorfismo de Nucleótido Simple , Humanos , Células K562 , Sitios de Carácter Cuantitativo , Células THP-1
12.
Sci Rep ; 10(1): 814, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31964908

RESUMEN

Myrf is a membrane-bound transcription factor that plays a key role in various biological processes. The Intramolecular Chaperone Auto-processing (ICA) domain of Myrf forms a homo-trimer, which carries out the auto-cleavage of Myrf. The ICA homo-trimer-mediated auto-cleavage of Myrf is a prerequisite for its transcription factor function in the nucleus. Recent exome sequencing studies have implicated two MYRF ICA domain mutations (V679A and R695H) in a novel syndromic form of birth defects. It remains unknown whether and how the two mutations impact the transcription factor function of Myrf and, more importantly, how they are pathogenic for congenital anomalies. Here, we show that V679A and R695H cripple the ICA domain, blocking the auto-cleavage of Myrf. Consequently, Myrf-V679A and Myrf-R695H do not exhibit any transcriptional activity. Molecular modeling suggests that V679A and R695H abrogate the auto-cleavage function of the ICA homo-trimer by destabilizing its homo-trimeric assembly. We also found that the ICA homo-trimer can tolerate one copy of Myrf-V679A or Myrf-R695H for its auto-cleavage function, indicating that V679A and R695H are not dominant negatives. Thus, if V679A and R695H in a heterozygous state caused birth defects, it would be via haploinsufficiency of MYRF.


Asunto(s)
Hernias Diafragmáticas Congénitas/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Células HEK293 , Haploinsuficiencia , Humanos , Proteínas de la Membrana/química , Modelos Moleculares , Mutación Missense , Conformación Proteica , Dominios Proteicos , Factores de Transcripción/química
13.
Cell Rep ; 32(11): 108147, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32937136

RESUMEN

PRC2 creates the repressive mark histone H3 Lys27 trimethylation. Although PRC2 is involved in various biological processes, its role in glial development remains ambiguous. Here, we show that PRC2 is required for oligodendrocyte (OL) differentiation and myelination, but not for OL precursor formation. PRC2-deficient OL lineage cells differentiate into OL precursors, but they fail to trigger the molecular program for myelination, highlighting that PRC2 is essential for directing the differentiation timing of OL precursors. PRC2 null OL lineage cells aberrantly induce Notch pathway genes and acquire astrocytic features. The repression of the Notch pathway restores the myelination program and inhibits abnormal astrocytic differentiation in the PRC2-deficient OL lineage, indicating that Notch is a major target of PRC2. Altogether, our studies propose a specific action of PRC2 as a novel gatekeeper that determines the glial fate choice and the timing of OL lineage progression and myelination by impinging on the Notch pathway.


Asunto(s)
Astrocitos/citología , Astrocitos/metabolismo , Linaje de la Célula , Oligodendroglía/citología , Oligodendroglía/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Animales , Diferenciación Celular , Pollos , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Metilación , Ratones , Vaina de Mielina/metabolismo , Factores de Transcripción NFI/metabolismo , Médula Espinal/citología , Médula Espinal/ultraestructura , Células Madre/citología , Células Madre/metabolismo , Vía de Señalización Wnt
14.
BMC Bioinformatics ; 10: 419, 2009 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-20003442

RESUMEN

BACKGROUND: Protein-protein interactions underlie many important biological processes. Computational prediction methods can nicely complement experimental approaches for identifying protein-protein interactions. Recently, a unique category of sequence-based prediction methods has been put forward--unique in the sense that it does not require homologous protein sequences. This enables it to be universally applicable to all protein sequences unlike many of previous sequence-based prediction methods. If effective as claimed, these new sequence-based, universally applicable prediction methods would have far-reaching utilities in many areas of biology research. RESULTS: Upon close survey, I realized that many of these new methods were ill-tested. In addition, newer methods were often published without performance comparison with previous ones. Thus, it is not clear how good they are and whether there are significant performance differences among them. In this study, I have implemented and thoroughly tested 4 different methods on large-scale, non-redundant data sets. It reveals several important points. First, significant performance differences are noted among different methods. Second, data sets typically used for training prediction methods appear significantly biased, limiting the general applicability of prediction methods trained with them. Third, there is still ample room for further developments. In addition, my analysis illustrates the importance of complementary performance measures coupled with right-sized data sets for meaningful benchmark tests. CONCLUSIONS: The current study reveals the potentials and limits of the new category of sequence-based protein-protein interaction prediction methods, which in turn provides a firm ground for future endeavours in this important area of contemporary bioinformatics.


Asunto(s)
Biología Computacional/métodos , Mapeo de Interacción de Proteínas/métodos , Proteínas/química , Análisis de Secuencia de Proteína/métodos , Secuencia de Aminoácidos , Sitios de Unión , Bases de Datos de Proteínas , Alineación de Secuencia
15.
Bioinformatics ; 24(16): 1819-20, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18544549

RESUMEN

UNLABELLED: To be fully functional, membrane proteins should not only fold, but also get inserted into the membrane, which is mediated by the Sec61 translocon. Recent experimental studies have attempted to elucidate how the Sec61 translocon accomplishes this delicate task by measuring the translocon-mediated membrane insertion free energies of 357 systematically designed peptides. On the basis of this data set, we have developed MINS2, a novel sequence-based computational method for predicting the membrane insertion free energies of protein sequences. A benchmark analysis of MINS2 shows that MINS2 signi.cantly outperforms previously proposed methods. Importantly, the application of MINS2 to known membrane protein structures shows that a better prediction of membrane insertion free energies does not lead to a better prediction of transmembrane segments of polytopic membrane proteins. AVAILABILITY: A web server for MINS2 is publicly available at http://service.bioinformatik.uni-saarland.de/mins. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Membrana Celular/química , Proteínas de la Membrana/química , Análisis de Secuencia de Proteína/métodos , Programas Informáticos , Secuencia de Aminoácidos , Sitios de Unión , Datos de Secuencia Molecular , Unión Proteica , Canales de Translocación SEC
16.
Bioinformatics ; 24(10): 1271-7, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18388143

RESUMEN

MOTIVATION: Helical membrane proteins (HMPs) play crucial roles in a variety of cellular processes. Unlike water-soluble proteins, HMPs need not only to fold but also get inserted into the membrane to be fully functional. This process of membrane insertion is mediated by the translocon complex. Thus, it is of great interest to develop computational methods for predicting the translocon-mediated membrane insertion free energies of protein sequences. RESULT: We have developed Membrane Insertion (MINS), a novel sequence-based computational method for predicting the membrane insertion free energies of protein sequences. A benchmark test gives a correlation coefficient of 0.74 between predicted and observed free energies for 357 known cases, which corresponds to a mean unsigned error of 0.41 kcal/mol. These results are significantly better than those obtained by traditional hydropathy analysis. Moreover, the ability of MINS to reasonably predict membrane insertion free energies of protein sequences allows for effective identification of transmembrane (TM) segments. Subsequently, MINS was applied to predict the membrane insertion free energies of 316 TM segments found in known structures. An in-depth analysis of the predicted free energies reveals a number of interesting findings about the biogenesis and structural stability of HMPs. AVAILABILITY: A web server for MINS is available at http://service.bioinformatik.uni-saarland.de/mins


Asunto(s)
Membrana Celular/química , Proteínas de la Membrana/química , Modelos Químicos , Análisis de Secuencia de Proteína/métodos , Secuencia de Aminoácidos , Simulación por Computador , Transferencia de Energía , Datos de Secuencia Molecular , Relación Estructura-Actividad , Termodinámica
17.
Front Mol Neurosci ; 12: 256, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31680860

RESUMEN

[This corrects the article DOI: 10.3389/fnmol.2019.00177.].

18.
Sci Rep ; 9(1): 11043, 2019 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-31363138

RESUMEN

Mapping enhancers to genes is a fundamental goal of modern biology. We have developed an innovative strategy that maps enhancers to genes in a principled manner. We illustrate its power by applying it to Myrf. Despite being a master regulator of oligodendrocytes, oligodendrocyte enhancers governing Myrf expression remain elusive. Since chromatin conformation capture studies have shown that a gene and its enhancer tend to be found in the same topologically associating domain (TAD), we started with the delineation of the Myrf TAD. A genome-wide map of putative oligodendrocyte enhancers uncovered 6 putative oligodendrocyte enhancers in the Myrf TAD, narrowing down the search space for Myrf enhancers from the entire genome to 6 loci in a principled manner. Epigenome editing experiments revealed that two of them govern Myrf expression for oligodendrocyte development. Our new method is simple, principled, and powerful, providing a systematic way to find enhancers that regulate the expression of a gene of interest. Since it can be applied to most cell types, it would greatly facilitate our effort to unravel transcriptional regulatory networks of diverse cell types.


Asunto(s)
Elementos de Facilitación Genéticos , Sitios Genéticos , Análisis de Secuencia de ADN/métodos , Factores de Transcripción/genética , Animales , Células Cultivadas , Ensamble y Desensamble de Cromatina , Regulación del Desarrollo de la Expresión Génica , Ratones , Oligodendroglía/citología , Oligodendroglía/metabolismo
19.
Bioinformatics ; 23(6): 701-8, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17237049

RESUMEN

Helical membrane proteins (HMPs) play a crucial role in diverse physiological processes. Given the difficulty in determining their structures by experimental techniques, it is desired to develop computational methods for predicting the burial status of transmembrane residues. Deriving a propensity scale for the 20 amino acids to be exposed to the lipid bilayer from known structures is central to developing such methods. A fundamental problem in this regard is what would be the optimal way of deriving propensity scales. Here, we show that this problem can be reformulated such that an optimal scale is straightforwardly obtained in an analytical fashion. The derived scale favorably compares with others in terms of both algorithmic optimality and practical prediction accuracy. It also allows interesting insights into the structural organization of HMPs. Furthermore, the presented approach can be applied to other bioinformatics problems of HMPs, too. All the data sets and programs used in the study and detailed primary results are available upon request.


Asunto(s)
Algoritmos , Aminoácidos/química , Inteligencia Artificial , Membrana Celular/química , Proteínas de la Membrana/química , Modelos Químicos , Análisis de Secuencia de Proteína/métodos , Secuencia de Aminoácidos , Simulación por Computador , Datos de Secuencia Molecular
20.
Sci Rep ; 8(1): 13075, 2018 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-30166609

RESUMEN

Myrf is a newly discovered membrane-bound transcription factor that plays an essential role in as diverse organisms as human, worm, and slime mold. Myrf is generated as a type-II membrane protein in the endoplasmic reticulum (ER). It forms homo-oligomers to undergo auto-cleavage that releases Myrf N-terminal fragment from the ER membrane as a homo-trimer. The homo-trimer of Myrf N-terminal fragments enters the nucleus and binds the Myrf motif to activate transcription. Despite its prominent role as a transcriptional activator, little is known about the transactivation domain of Myrf. Here, we report that the N-terminal-most (NTM) domain of Myrf is required for transcriptional activity and, when fused to a Gal4 fragment, enables it to activate transcription. The transactivation function of the NTM domain did not require homo-trimerization. We also discovered that the NTM domain can be sumoylated at three lysine residues (K123, K208, and K276), with K276 serving as the main acceptor. K276 sumoylation repressed the transactivation function of the NTM domain without affecting the stability or nuclear localization of Myrf N-terminal fragment. In sum, this study identifies the NTM domain as the transactivation domain of Myrf and the potential regulatory impact of its K276 sumoylation.


Asunto(s)
Pleiotropía Genética , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Núcleo Celular/metabolismo , ADN/metabolismo , Lisina/metabolismo , Ratones , Unión Proteica , Dominios Proteicos , Sumoilación , Factores de Transcripción/genética , Transcripción Genética , Activación Transcripcional/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA