Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Brief Bioinform ; 24(5)2023 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-37649383

RESUMEN

Single-cell high-throughput chromatin conformation capture technologies (scHi-C) has been used to map chromatin spatial organization in complex tissues. However, computational tools to detect differential chromatin contacts (DCCs) from scHi-C datasets in development and through disease pathogenesis are still lacking. Here, we present SnapHiC-D, a computational pipeline to identify DCCs between two scHi-C datasets. Compared to methods designed for bulk Hi-C data, SnapHiC-D detects DCCs with high sensitivity and accuracy. We used SnapHiC-D to identify cell-type-specific chromatin contacts at 10 Kb resolution in mouse hippocampal and human prefrontal cortical tissues, demonstrating that DCCs detected in the hippocampal and cortical cell types are generally associated with cell-type-specific gene expression patterns and epigenomic features. SnapHiC-D is freely available at https://github.com/HuMingLab/SnapHiC-D.


Asunto(s)
Cromatina , Epigenómica , Humanos , Animales , Ratones , Cromatina/genética , Hipocampo
2.
Cell Rep ; 42(8): 112896, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37505983

RESUMEN

The impact of chromosomal inversions on human brain morphology remains underexplored. We studied 35 common inversions classified from genotypes of 33,018 adults with European ancestry. The inversions at 2p22.3, 16p11.2, and 17q21.31 reach genome-wide significance, followed by 8p23.1 and 6p21.33, in their association with cortical and subcortical morphology. The 17q21.31, 8p23.1, and 16p11.2 regions comprise the LRRC37, OR7E, and NPIP duplicated gene families. We find the 17q21.31 MAPT inversion region, known for harboring neurological risk, to be the most salient locus among common variants for shaping and patterning the cortex. Overall, we observe the inverted orientations decreasing brain size, with the exception that the 2p22.3 inversion is associated with increased subcortical volume and the 8p23.1 inversion is associated with increased motor cortex. These significant inversions are in the genomic hotspots of neuropsychiatric loci. Our findings are generalizable to 3,472 children and demonstrate inversions as essential genetic variation to understand human brain phenotypes.


Asunto(s)
Inversión Cromosómica , Polimorfismo Genético , Adulto , Niño , Humanos , Inversión Cromosómica/genética , Encéfalo
3.
Commun Biol ; 6(1): 435, 2023 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-37081156

RESUMEN

Topologically associating domain (TAD) boundaries partition the genome into distinct regulatory territories. Anecdotal evidence suggests that their disruption may interfere with normal gene expression and cause disease phenotypes1-3, but the overall extent to which this occurs remains unknown. Here we demonstrate that targeted deletions of TAD boundaries cause a range of disruptions to normal in vivo genome function and organismal development. We used CRISPR genome editing in mice to individually delete eight TAD boundaries (11-80 kb in size) from the genome. All deletions examined resulted in detectable molecular or organismal phenotypes, which included altered chromatin interactions or gene expression, reduced viability, and anatomical phenotypes. We observed changes in local 3D chromatin architecture in 7 of 8 (88%) cases, including the merging of TADs and altered contact frequencies within TADs adjacent to the deleted boundary. For 5 of 8 (63%) loci examined, boundary deletions were associated with increased embryonic lethality or other developmental phenotypes. For example, a TAD boundary deletion near Smad3/Smad6 caused complete embryonic lethality, while a deletion near Tbx5/Lhx5 resulted in a severe lung malformation. Our findings demonstrate the importance of TAD boundary sequences for in vivo genome function and reinforce the critical need to carefully consider the potential pathogenicity of noncoding deletions affecting TAD boundaries in clinical genetics screening.


Asunto(s)
Cromatina , Genoma , Animales , Ratones , Cromatina/genética , Fenotipo
4.
Cell Res ; 32(11): 1008-1021, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36207411

RESUMEN

Loss of heterochromatin has been implicated as a cause of pre-mature aging and age-associated decline in organ functions in mammals; however, the specific cell types and gene loci affected by this type of epigenetic change have remained unclear. To address this knowledge gap, we probed chromatin accessibility at single-cell resolution in the brains, hearts, skeletal muscles, and bone marrows from young, middle-aged, and old mice, and assessed age-associated changes at 353,126 candidate cis-regulatory elements (cCREs) across 32 major cell types. Unexpectedly, we detected increased chromatin accessibility within specific heterochromatin domains in old mouse excitatory neurons. The gain of chromatin accessibility at these genomic loci was accompanied by the cell-type-specific loss of heterochromatin and activation of LINE1 elements. Immunostaining further confirmed the loss of the heterochromatin mark H3K9me3 in the excitatory neurons but not in inhibitory neurons or glial cells. Our results reveal the cell-type-specific changes in chromatin landscapes in old mice and shed light on the scope of heterochromatin loss in mammalian aging.


Asunto(s)
Epigenoma , Heterocromatina , Ratones , Animales , Cromatina , Neuronas , Encéfalo , Mamíferos/genética
5.
Nat Cardiovasc Res ; 1(9): 830-843, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36817700

RESUMEN

The heart, a vital organ which is first to develop, has adapted its size, structure and function in order to accommodate the circulatory demands for a broad range of animals. Although heart development is controlled by a relatively conserved network of transcriptional/chromatin regulators, how the human heart has evolved species-specific features to maintain adequate cardiac output and function remains to be defined. Here, we show through comparative epigenomic analysis the identification of enhancers and promoters that have gained activity in humans during cardiogenesis. These cis-regulatory elements (CREs) are associated with genes involved in heart development and function, and may account for species-specific differences between human and mouse hearts. Supporting these findings, genetic variants that are associated with human cardiac phenotypic/disease traits, particularly those differing between human and mouse, are enriched in human-gained CREs. During early stages of human cardiogenesis, these CREs are also gained within genomic loci of transcriptional regulators, potentially expanding their role in human heart development. In particular, we discovered that gained enhancers in the locus of the early human developmental regulator ZIC3 are selectively accessible within a subpopulation of mesoderm cells which exhibits cardiogenic potential, thus possibly extending the function of ZIC3 beyond its conserved left-right asymmetry role. Genetic deletion of these enhancers identified a human gained enhancer that was required for not only ZIC3 and early cardiac gene expression at the mesoderm stage but also cardiomyocyte differentiation. Overall, our results illuminate how human gained CREs may contribute to human-specific cardiac attributes, and provide insight into how transcriptional regulators may gain cardiac developmental roles through the evolutionary acquisition of enhancers.

6.
Nat Methods ; 18(9): 1056-1059, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34446921

RESUMEN

Single-cell Hi-C (scHi-C) analysis has been increasingly used to map chromatin architecture in diverse tissue contexts, but computational tools to define chromatin loops at high resolution from scHi-C data are still lacking. Here, we describe Single-Nucleus Analysis Pipeline for Hi-C (SnapHiC), a method that can identify chromatin loops at high resolution and accuracy from scHi-C data. Using scHi-C data from 742 mouse embryonic stem cells, we benchmark SnapHiC against a number of computational tools developed for mapping chromatin loops and interactions from bulk Hi-C. We further demonstrate its use by analyzing single-nucleus methyl-3C-seq data from 2,869 human prefrontal cortical cells, which uncovers cell type-specific chromatin loops and predicts putative target genes for noncoding sequence variants associated with neuropsychiatric disorders. Our results indicate that SnapHiC could facilitate the analysis of cell type-specific chromatin architecture and gene regulatory programs in complex tissues.


Asunto(s)
Cromatina/química , Biología Computacional/métodos , Análisis de la Célula Individual/métodos , Algoritmos , Animales , Cromatina/genética , Secuenciación de Inmunoprecipitación de Cromatina , Visualización de Datos , Bases de Datos Factuales , Expresión Génica , Humanos , Trastornos Mentales/genética , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/fisiología , Polimorfismo de Nucleótido Simple , Corteza Prefrontal/citología , Reproducibilidad de los Resultados , Análisis de Secuencia de ADN/métodos
7.
Mol Cell ; 81(16): 3294-3309.e12, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34293321

RESUMEN

Temperature is a variable component of the environment, and all organisms must deal with or adapt to temperature change. Acute temperature change activates cellular stress responses, resulting in refolding or removal of damaged proteins. However, how organisms adapt to long-term temperature change remains largely unexplored. Here we report that budding yeast responds to long-term high temperature challenge by switching from chaperone induction to reduction of temperature-sensitive proteins and re-localizing a portion of its proteome. Surprisingly, we also find that many proteins adopt an alternative conformation. Using Fet3p as an example, we find that the temperature-dependent conformational difference is accompanied by distinct thermostability, subcellular localization, and, importantly, cellular functions. We postulate that, in addition to the known mechanisms of adaptation, conformational plasticity allows some polypeptides to acquire new biophysical properties and functions when environmental change endures.


Asunto(s)
Adaptación Fisiológica/genética , Proteoma/genética , Estrés Fisiológico/genética , Transcriptoma/genética , Aclimatación/genética , Animales , Exposición a Riesgos Ambientales/efectos adversos , Regulación Fúngica de la Expresión Génica/genética , Calor/efectos adversos , Saccharomycetales/genética
8.
Sci Adv ; 7(20)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33990324

RESUMEN

Misregulated gene expression in human hearts can result in cardiovascular diseases that are leading causes of mortality worldwide. However, the limited information on the genomic location of candidate cis-regulatory elements (cCREs) such as enhancers and promoters in distinct cardiac cell types has restricted the understanding of these diseases. Here, we defined >287,000 cCREs in the four chambers of the human heart at single-cell resolution, which revealed cCREs and candidate transcription factors associated with cardiac cell types in a region-dependent manner and during heart failure. We further found cardiovascular disease-associated genetic variants enriched within these cCREs including 38 candidate causal atrial fibrillation variants localized to cardiomyocyte cCREs. Additional functional studies revealed that two of these variants affect a cCRE controlling KCNH2/HERG expression and action potential repolarization. Overall, this atlas of human cardiac cCREs provides the foundation for illuminating cell type-specific gene regulation in human hearts during health and disease.


Asunto(s)
Corazón , Secuencias Reguladoras de Ácidos Nucleicos , Humanos , Regiones Promotoras Genéticas , Secuencias Reguladoras de Ácidos Nucleicos/genética , Factores de Transcripción/metabolismo
9.
Nat Genet ; 53(7): 1064-1074, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34002095

RESUMEN

Insulators play a critical role in spatiotemporal gene regulation in animals. The evolutionarily conserved CCCTC-binding factor (CTCF) is required for insulator function in mammals, but not all of its binding sites act as insulators. Here we explore the sequence requirements of CTCF-mediated transcriptional insulation using a sensitive insulator reporter in mouse embryonic stem cells. We find that insulation potency depends on the number of CTCF-binding sites in tandem. Furthermore, CTCF-mediated insulation is dependent on upstream flanking sequences at its binding sites. CTCF-binding sites at topologically associating domain boundaries are more likely to function as insulators than those outside topologically associating domain boundaries, independently of binding strength. We demonstrate that insulators form local chromatin domain boundaries and weaken enhancer-promoter contacts. Taken together, our results provide genetic, molecular and structural evidence connecting chromatin topology to the action of insulators in the mammalian genome.


Asunto(s)
Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/metabolismo , Cromatina/genética , Cromatina/metabolismo , Regulación de la Expresión Génica , Transcripción Genética , Animales , Sitios de Unión , Factor de Unión a CCCTC/química , Elementos de Facilitación Genéticos , Humanos , Elementos Aisladores , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Regiones Promotoras Genéticas , Unión Proteica
10.
Nat Commun ; 12(1): 1337, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33637727

RESUMEN

Identification of the cis-regulatory elements controlling cell-type specific gene expression patterns is essential for understanding the origin of cellular diversity. Conventional assays to map regulatory elements via open chromatin analysis of primary tissues is hindered by sample heterogeneity. Single cell analysis of accessible chromatin (scATAC-seq) can overcome this limitation. However, the high-level noise of each single cell profile and the large volume of data pose unique computational challenges. Here, we introduce SnapATAC, a software package for analyzing scATAC-seq datasets. SnapATAC dissects cellular heterogeneity in an unbiased manner and map the trajectories of cellular states. Using the Nyström method, SnapATAC can process data from up to a million cells. Furthermore, SnapATAC incorporates existing tools into a comprehensive package for analyzing single cell ATAC-seq dataset. As demonstration of its utility, SnapATAC is applied to 55,592 single-nucleus ATAC-seq profiles from the mouse secondary motor cortex. The analysis reveals ~370,000 candidate regulatory elements in 31 distinct cell populations in this brain region and inferred candidate cell-type specific transcriptional regulators.


Asunto(s)
Secuenciación de Inmunoprecipitación de Cromatina/métodos , Análisis de la Célula Individual/métodos , Animales , Cromatina , Biología Computacional , Epigenómica , Masculino , Ratones , Ratones Endogámicos C57BL , Corteza Motora , Análisis de Secuencia de ADN/métodos
11.
Nat Methods ; 18(3): 283-292, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33589836

RESUMEN

Genome-wide profiling of histone modifications can reveal not only the location and activity state of regulatory elements, but also the regulatory mechanisms involved in cell-type-specific gene expression during development and disease pathology. Conventional assays to profile histone modifications in bulk tissues lack single-cell resolution. Here we describe an ultra-high-throughput method, Paired-Tag, for joint profiling of histone modifications and transcriptome in single cells to produce cell-type-resolved maps of chromatin state and transcriptome in complex tissues. We used this method to profile five histone modifications jointly with transcriptome in the adult mouse frontal cortex and hippocampus. Integrative analysis of the resulting maps identified distinct groups of genes subject to divergent epigenetic regulatory mechanisms. Our single-cell multiomics approach enables comprehensive analysis of chromatin state and gene regulation in complex tissues and characterization of gene regulatory programs in the constituent cell types.


Asunto(s)
Lóbulo Frontal/metabolismo , Regulación de la Expresión Génica/genética , Hipocampo/metabolismo , Código de Histonas/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Animales , Línea Celular Tumoral , Cromatina/metabolismo , Epigénesis Genética/genética , Lóbulo Frontal/citología , Perfilación de la Expresión Génica , Células HeLa , Hipocampo/citología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Procesamiento Proteico-Postraduccional , Análisis de la Célula Individual , Transcriptoma/genética
14.
Nature ; 583(7818): 744-751, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32728240

RESUMEN

The Encyclopedia of DNA Elements (ENCODE) project has established a genomic resource for mammalian development, profiling a diverse panel of mouse tissues at 8 developmental stages from 10.5 days after conception until birth, including transcriptomes, methylomes and chromatin states. Here we systematically examined the state and accessibility of chromatin in the developing mouse fetus. In total we performed 1,128 chromatin immunoprecipitation with sequencing (ChIP-seq) assays for histone modifications and 132 assay for transposase-accessible chromatin using sequencing (ATAC-seq) assays for chromatin accessibility across 72 distinct tissue-stages. We used integrative analysis to develop a unified set of chromatin state annotations, infer the identities of dynamic enhancers and key transcriptional regulators, and characterize the relationship between chromatin state and accessibility during developmental gene regulation. We also leveraged these data to link enhancers to putative target genes and demonstrate tissue-specific enrichments of sequence variants associated with disease in humans. The mouse ENCODE data sets provide a compendium of resources for biomedical researchers and achieve, to our knowledge, the most comprehensive view of chromatin dynamics during mammalian fetal development to date.


Asunto(s)
Cromatina/genética , Cromatina/metabolismo , Conjuntos de Datos como Asunto , Desarrollo Fetal/genética , Histonas/metabolismo , Anotación de Secuencia Molecular , Secuencias Reguladoras de Ácidos Nucleicos/genética , Animales , Cromatina/química , Secuenciación de Inmunoprecipitación de Cromatina , Enfermedad/genética , Elementos de Facilitación Genéticos/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Variación Genética , Histonas/química , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos/genética , Reproducibilidad de los Resultados , Transposasas/metabolismo
15.
Head Neck ; 42(9): 2375-2389, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32406560

RESUMEN

BACKGROUND: Human papillomavirus (HPV) oncogenes E6, E7, and shorter isoforms of E6 (E6*) are known carcinogenic factors in head and neck squamous cell carcinoma (HNSCC). Little is known regarding E6* functions. METHODS: We analyzed RNA-seq data from 68 HNSCC HPV type 16-positive tumors to determine host genes and pathways associated with E6+E7 expression (E6E7) or the percent of full-length E6 (E6%FL). Influence scores of E6E7 and E6%FL were used to test for associations with clinical variables. RESULTS: For E6E7, we recapitulated all major known affected pathways and revealed additional pathways. E6%FL was found to affect mitochondrial processes, and E6%FL influence score was significantly associated with overall survival and tumor size. CONCLUSIONS: HPV E6E7 and E6* result in extensive, dose-dependent compensatory effects and dysregulation of key cancer pathways. The switch from E6 to E6* promotes oxidative phosphorylation, larger tumor size, and worse prognosis, potentially serving as a prognostic factor for HPV-positive HNSCC.


Asunto(s)
Alphapapillomavirus , Neoplasias de Cabeza y Cuello , Proteínas Oncogénicas Virales , Infecciones por Papillomavirus , Carcinógenos , Femenino , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/virología , Humanos , Masculino , Proteínas Oncogénicas Virales/genética , Oncogenes , Papillomaviridae/genética , Proteínas E7 de Papillomavirus/genética , Infecciones por Papillomavirus/genética , Proteínas Represoras/genética , Transcriptoma
16.
Nat Commun ; 11(1): 1122, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-32111823

RESUMEN

Heart failure is a major public health problem affecting over 23 million people worldwide. In this study, we present the results of a large scale meta-analysis of heart failure GWAS and replication in a comparable sized cohort to identify one known and two novel loci associated with heart failure. Heart failure sub-phenotyping shows that a new locus in chromosome 1 is associated with left ventricular adverse remodeling and clinical heart failure, in response to different initial cardiac muscle insults. Functional characterization and fine-mapping of that locus reveal a putative causal variant in a cardiac muscle specific regulatory region activated during cardiomyocyte differentiation that binds to the ACTN2 gene, a crucial structural protein inside the cardiac sarcolemma (Hi-C interaction p-value = 0.00002). Genome-editing in human embryonic stem cell-derived cardiomyocytes confirms the influence of the identified regulatory region in the expression of ACTN2. Our findings extend our understanding of biological mechanisms underlying heart failure.


Asunto(s)
Actinina/genética , Predisposición Genética a la Enfermedad/genética , Insuficiencia Cardíaca/genética , Sistema del Grupo Sanguíneo ABO/genética , Fibrilación Atrial/genética , Cromosomas Humanos Par 1 , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Insuficiencia Cardíaca/patología , Células Madre Embrionarias Humanas/citología , Humanos , Enfermedades Musculoesqueléticas/genética , Miocitos Cardíacos/citología , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo
17.
Cancer Cell ; 37(4): 584-598.e11, 2020 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-32220301

RESUMEN

Metastatic prostate cancer is characterized by recurrent genomic copy number alterations that are presumed to contribute to resistance to hormone therapy. We identified CHD1 loss as a cause of antiandrogen resistance in an in vivo small hairpin RNA (shRNA) screen of 730 genes deleted in prostate cancer. ATAC-seq and RNA-seq analyses showed that CHD1 loss resulted in global changes in open and closed chromatin with associated transcriptomic changes. Integrative analysis of this data, together with CRISPR-based functional screening, identified four transcription factors (NR3C1, POU3F2, NR2F1, and TBX2) that contribute to antiandrogen resistance, with associated activation of non-luminal lineage programs. Thus, CHD1 loss results in chromatin dysregulation, thereby establishing a state of transcriptional plasticity that enables the emergence of antiandrogen resistance through heterogeneous mechanisms.


Asunto(s)
Antagonistas de Andrógenos/farmacología , Cromatina/genética , ADN Helicasas/antagonistas & inhibidores , Proteínas de Unión al ADN/antagonistas & inhibidores , Resistencia a Antineoplásicos/genética , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , ARN Interferente Pequeño/genética , Receptores Androgénicos/química , Animales , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Cromatina/metabolismo , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Ensayos Analíticos de Alto Rendimiento , Humanos , Masculino , Ratones , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptores Androgénicos/genética , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
18.
J Paediatr Child Health ; 56(2): 231-236, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31408250

RESUMEN

AIM: To examine the association of life-style factors, including second-hand smoke, with dental caries among 3-year-old children in Wuxi, China. METHODS: A multi-stage stratified random cluster sampling method was used, and 283 children were recruited. The prevalence of dental caries was 29.3% (83/283). RESULTS: Univariate analysis indicated that the possible related factors of dental caries included sleep duration, interest in snacks, candy, exposure to second-hand smoke and weight of birth (all P < 0.05). Meanwhile, multivariate logistic regression analysis suggested that children who had used fluoride were less susceptible to dental caries than those who had not used fluoride before (P < 0.05). Moreover, the risk of dental caries in children who were very interested in snacks was greater than those with little interest in snacks (P < 0.05). CONCLUSIONS: Life-style behaviours are crucial factors and should attract enough attention. There might be a potential negative effect of second-hand smoke on the deciduous caries, but it still requires further studies. A co-ordinated effort by health-care providers, policymakers and health institutions has successfully improved children's oral health and the awareness of hygiene knowledge among citizens in Wuxi city.


Asunto(s)
Caries Dental , Contaminación por Humo de Tabaco , Preescolar , China/epidemiología , Estudios Transversales , Índice CPO , Caries Dental/epidemiología , Caries Dental/etiología , Humanos , Estilo de Vida , Prevalencia , Factores de Riesgo , Contaminación por Humo de Tabaco/efectos adversos
19.
J Neurosci ; 39(43): 8457-8470, 2019 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-31492772

RESUMEN

The degeneration of injured axons involves a self-destruction pathway whose components and mechanism are not fully understood. Here, we report a new regulator of axonal resilience. The transmembrane protein Raw is cell autonomously required for the degeneration of injured axons, dendrites, and synapses in Drosophila melanogaster In both male and female raw hypomorphic mutant or knock-down larvae, the degeneration of injured axons, dendrites, and synapses from motoneurons and sensory neurons is strongly inhibited. This protection is insensitive to reduction in the levels of the NAD+ synthesis enzyme Nmnat (nicotinamide mononucleotide adenylyl transferase), but requires the c-Jun N-terminal kinase (JNK) mitogen-activated protein (MAP) kinase and the transcription factors Fos and Jun (AP-1). Although these factors were previously known to function in axonal injury signaling and regeneration, Raw's function can be genetically separated from other axonal injury responses: Raw does not modulate JNK-dependent axonal injury signaling and regenerative responses, but instead restrains a protective pathway that inhibits the degeneration of axons, dendrites, and synapses. Although protection in raw mutants requires JNK, Fos, and Jun, JNK also promotes axonal degeneration. These findings suggest the existence of multiple independent pathways that share modulation by JNK, Fos, and Jun that influence how axons respond to stress and injury.SIGNIFICANCE STATEMENT Axonal degeneration is a major feature of neuropathies and nerve injuries and occurs via a cell autonomous self-destruction pathway whose mechanism is poorly understood. This study reports the identification of a new regulator of axonal degeneration: the transmembrane protein Raw. Raw regulates a cell autonomous nuclear signaling pathway whose yet unknown downstream effectors protect injured axons, dendrites, and synapses from degenerating. These findings imply that the susceptibility of axons to degeneration is strongly regulated in neurons. Future understanding of the cellular pathway regulated by Raw, which engages the c-Jun N-terminal kinase (JNK) mitogen-activated protein (MAP) kinase and Fos and Jun transcription factors, may suggest new strategies to increase the resiliency of axons in debilitating neuropathies.


Asunto(s)
Axones/metabolismo , Proteínas del Citoesqueleto/metabolismo , Dendritas/metabolismo , Proteínas de Drosophila/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Degeneración Nerviosa/metabolismo , Animales , Animales Modificados Genéticamente , Axones/patología , Proteínas del Citoesqueleto/genética , Dendritas/patología , Proteínas de Drosophila/genética , Drosophila melanogaster , Femenino , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Degeneración Nerviosa/patología , Sinapsis/metabolismo
20.
Nat Methods ; 16(10): 991-993, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31384045

RESUMEN

We report a molecular assay, Methyl-HiC, that can simultaneously capture the chromosome conformation and DNA methylome in a cell. Methyl-HiC reveals coordinated DNA methylation status between distal genomic segments that are in spatial proximity in the nucleus, and delineates heterogeneity of both the chromatin architecture and DNA methylome in a mixed population. It enables simultaneous characterization of cell-type-specific chromatin organization and epigenome in complex tissues.


Asunto(s)
Cromatina/metabolismo , Metilación de ADN , Análisis de la Célula Individual/métodos , Animales , Islas de CpG , Conjuntos de Datos como Asunto , Humanos , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...