Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell ; 186(26): 5840-5858.e36, 2023 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-38134876

RESUMEN

Short tandem repeat (STR) instability causes transcriptional silencing in several repeat expansion disorders. In fragile X syndrome (FXS), mutation-length expansion of a CGG STR represses FMR1 via local DNA methylation. Here, we find megabase-scale H3K9me3 domains on autosomes and encompassing FMR1 on the X chromosome in FXS patient-derived iPSCs, iPSC-derived neural progenitors, EBV-transformed lymphoblasts, and brain tissue with mutation-length CGG expansion. H3K9me3 domains connect via inter-chromosomal interactions and demarcate severe misfolding of TADs and loops. They harbor long synaptic genes replicating at the end of S phase, replication-stress-induced double-strand breaks, and STRs prone to stepwise somatic instability. CRISPR engineering of the mutation-length CGG to premutation length reverses H3K9me3 on the X chromosome and multiple autosomes, refolds TADs, and restores gene expression. H3K9me3 domains can also arise in normal-length iPSCs created with perturbations linked to genome instability, suggesting their relevance beyond FXS. Our results reveal Mb-scale heterochromatinization and trans interactions among loci susceptible to instability.


Asunto(s)
Síndrome del Cromosoma X Frágil , Humanos , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Expansión de Repetición de Trinucleótido , Metilación de ADN , Mutación , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo
2.
Cell ; 175(1): 224-238.e15, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30173918

RESUMEN

More than 25 inherited human disorders are caused by the unstable expansion of repetitive DNA sequences termed short tandem repeats (STRs). A fundamental unresolved question is why some STRs are susceptible to pathologic expansion, whereas thousands of repeat tracts across the human genome are relatively stable. Here, we discover that nearly all disease-associated STRs (daSTRs) are located at boundaries demarcating 3D chromatin domains. We identify a subset of boundaries with markedly higher CpG island density compared to the rest of the genome. daSTRs specifically localize to ultra-high-density CpG island boundaries, suggesting they might be hotspots for epigenetic misregulation or topological disruption linked to STR expansion. Fragile X syndrome patients exhibit severe boundary disruption in a manner that correlates with local loss of CTCF occupancy and the degree of FMR1 silencing. Our data uncover higher-order chromatin architecture as a new dimension in understanding repeat expansion disorders.


Asunto(s)
Cromatina/genética , Repeticiones de Microsatélite/fisiología , Expansión de Repetición de Trinucleótido/fisiología , Adulto , Encéfalo/citología , Encéfalo/patología , Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/fisiología , Línea Celular , Cromatina/fisiología , Ensamble y Desensamble de Cromatina/genética , Ensamble y Desensamble de Cromatina/fisiología , Islas de CpG/genética , Islas de CpG/fisiología , ADN/genética , Enfermedad/etiología , Enfermedad/genética , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/fisiología , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Genoma Humano/genética , Humanos , Masculino , Repeticiones de Microsatélite/genética , Expansión de Repetición de Trinucleótido/genética
3.
Mol Cell ; 82(20): 3769-3780.e5, 2022 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-36182691

RESUMEN

Complex genomes show intricate organization in three-dimensional (3D) nuclear space. Current models posit that cohesin extrudes loops to form self-interacting domains delimited by the DNA binding protein CTCF. Here, we describe and quantitatively characterize cohesin-propelled, jet-like chromatin contacts as landmarks of loop extrusion in quiescent mammalian lymphocytes. Experimental observations and polymer simulations indicate that narrow origins of loop extrusion favor jet formation. Unless constrained by CTCF, jets propagate symmetrically for 1-2 Mb, providing an estimate for the range of in vivo loop extrusion. Asymmetric CTCF binding deflects the angle of jet propagation as experimental evidence that cohesin-mediated loop extrusion can switch from bi- to unidirectional and is controlled independently in both directions. These data offer new insights into the physiological behavior of in vivo cohesin-mediated loop extrusion and further our understanding of the principles that underlie genome organization.


Asunto(s)
Cromatina , Proteínas Cromosómicas no Histona , Animales , Cromatina/genética , Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Polímeros/metabolismo , Mamíferos/metabolismo , Cohesinas
4.
Immunity ; 52(2): 257-274.e11, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32049053

RESUMEN

Genetics is a major determinant of susceptibility to autoimmune disorders. Here, we examined whether genome organization provides resilience or susceptibility to sequence variations, and how this would contribute to the molecular etiology of an autoimmune disease. We generated high-resolution maps of linear and 3D genome organization in thymocytes of NOD mice, a model of type 1 diabetes (T1D), and the diabetes-resistant C57BL/6 mice. Multi-enhancer interactions formed at genomic regions harboring genes with prominent roles in T cell development in both strains. However, diabetes risk-conferring loci coalesced enhancers and promoters in NOD, but not C57BL/6 thymocytes. 3D genome mapping of NODxC57BL/6 F1 thymocytes revealed that genomic misfolding in NOD mice is mediated in cis. Moreover, immune cells infiltrating the pancreas of humans with T1D exhibited increased expression of genes located on misfolded loci in mice. Thus, genetic variation leads to altered 3D chromatin architecture and associated changes in gene expression that may underlie autoimmune pathology.


Asunto(s)
Cromatina/metabolismo , Diabetes Mellitus Tipo 1/genética , Predisposición Genética a la Enfermedad/genética , Timocitos/patología , Animales , Factor de Unión a CCCTC/metabolismo , Mapeo Cromosómico , Diabetes Mellitus Tipo 1/patología , Epigénesis Genética , Expresión Génica , Sitios Genéticos/genética , Variación Genética , Genoma/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Páncreas/patología , Secuencias Reguladoras de Ácidos Nucleicos
5.
Mol Cell ; 81(15): 3065-3081.e12, 2021 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-34297911

RESUMEN

The chromatin fiber folds into loops, but the mechanisms controlling loop extrusion are still poorly understood. Using super-resolution microscopy, we visualize that loops in intact nuclei are formed by a scaffold of cohesin complexes from which the DNA protrudes. RNA polymerase II decorates the top of the loops and is physically segregated from cohesin. Augmented looping upon increased loading of cohesin on chromosomes causes disruption of Lamin at the nuclear rim and chromatin blending, a homogeneous distribution of chromatin within the nucleus. Altering supercoiling via either transcription or topoisomerase inhibition counteracts chromatin blending, increases chromatin condensation, disrupts loop formation, and leads to altered cohesin distribution and mobility on chromatin. Overall, negative supercoiling generated by transcription is an important regulator of loop formation in vivo.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cromatina/química , Cromatina/genética , Proteínas Cromosómicas no Histona/metabolismo , Transcripción Genética/fisiología , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Línea Celular , Núcleo Celular/genética , Proteoglicanos Tipo Condroitín Sulfato/genética , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/genética , ADN-Topoisomerasas de Tipo I/genética , ADN-Topoisomerasas de Tipo I/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Laminas/genética , Laminas/metabolismo , ARN Polimerasa II/metabolismo , Imagen Individual de Molécula/métodos , Cohesinas
6.
Phys Biol ; 20(5)2023 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-37467757

RESUMEN

Eukaryotic chromosomes exhibit a hierarchical organization that spans a spectrum of length scales, ranging from sub-regions known as loops, which typically comprise hundreds of base pairs, to much larger chromosome territories that can encompass a few mega base pairs. Chromosome conformation capture experiments that involve high-throughput sequencing methods combined with microscopy techniques have enabled a new understanding of inter- and intra-chromosomal interactions with unprecedented details. This information also provides mechanistic insights on the relationship between genome architecture and gene expression. In this article, we review the recent findings on three-dimensional interactions among chromosomes at the compartment, topologically associating domain, and loop levels and the impact of these interactions on the transcription process. We also discuss current understanding of various biophysical processes involved in multi-layer structural organization of chromosomes. Then, we discuss the relationships between gene expression and genome structure from perturbative genome-wide association studies. Furthermore, for a better understanding of how chromosome architecture and function are linked, we emphasize the role of epigenetic modifications in the regulation of gene expression. Such an understanding of the relationship between genome architecture and gene expression can provide a new perspective on the range of potential future discoveries and therapeutic research.


Asunto(s)
Cromosomas , Estudio de Asociación del Genoma Completo , Genoma , Eucariontes/genética , Conformación Molecular , Cromatina/genética
7.
Methods ; 170: 48-60, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31252062

RESUMEN

Studies performed using Hi-C and other high-throughput whole-genome C-methods have demonstrated that 3D organization of eukaryotic genomes is functionally relevant. Unfortunately, ultra-deep sequencing of Hi-C libraries necessary to detect loop structures in large vertebrate genomes remains rather expensive. However, many studies are in fact aimed at determining the fine-scale 3D structure of comparatively small genomic regions up to several Mb in length. Such studies typically focus on the spatial structure of domains of coregulated genes, molecular mechanisms of loop formation, and interrogation of functional significance of GWAS-revealed polymorphisms. Therefore, a handful of molecular techniques based on Hi-C have been developed to address such issues. These techniques commonly rely on in-solution hybridization of Hi-C/3C-seq libraries with pools of biotinylated baits covering the region of interest, followed by deep sequencing of the enriched library. Here, we describe a new protocol of this kind, C-TALE (Chromatin TArget Ligation Enrichment). Preparation of hybridization probes from bacterial artificial chromosomes and an additional round of enrichment make C-TALE a cost-effective alternative to existing many-versus-all C-methods.


Asunto(s)
Mapeo Cromosómico/métodos , Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Animales , Biotinilación , Línea Celular , Cromatina/química , Cromatina/genética , Cromatina/aislamiento & purificación , Cromatina/metabolismo , Mapeo Cromosómico/economía , Cromosomas Artificiales Bacterianos/genética , ADN/genética , ADN/aislamiento & purificación , ADN/metabolismo , Biblioteca de Genes , Genómica/economía , Secuenciación de Nucleótidos de Alto Rendimiento/economía , Humanos , Conformación de Ácido Nucleico , Hibridación de Ácido Nucleico/métodos
8.
Cancer Sci ; 110(8): 2328-2336, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31228211

RESUMEN

Changes of nuclear localization of lineage-specific genes from a transcriptionally inert to permissive environment are a crucial step in establishing the identity of a cell. Noncoding RNA transcription-mediated genome folding and activation of target gene expression have been found in a variety of cell types. Noncoding RNA ThymoD (thymocyte differentiation factor) transcription at superenhancers is essential for mouse T-cell lineage commitment. The cessation of ThymoD transcription abolishes transcription-mediated demethylation, recruiting looping factors such as the cohesin complex, CCCTC-binding factor (CTCF), ultimately leading to the phenotype of severe combined immunodeficiency and T-cell leukemia/lymphoma. In this review, we describe the functional role of RNA polymerase II-mediated transcription at enhancers and in genome folding. We also highlight the involvement of faulty activation or suppression of enhancer transcription and enhancer-promoter interaction in cancer development.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Genoma/genética , Neoplasias/genética , ARN no Traducido/genética , Transcripción Genética/genética , Animales , Humanos , Regiones Promotoras Genéticas/genética
9.
Annu Rev Genomics Hum Genet ; 17: 17-43, 2016 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-27089971

RESUMEN

Genome function, replication, integrity, and propagation rely on the dynamic structural organization of chromosomes during the cell cycle. Genome folding in interphase provides regulatory segmentation for appropriate transcriptional control, facilitates ordered genome replication, and contributes to genome integrity by limiting illegitimate recombination. Here, we review recent high-resolution chromosome conformation capture and functional studies that have informed models of the spatial and regulatory compartmentalization of mammalian genomes, and discuss mechanistic models for how CTCF and cohesin control the functional architecture of mammalian chromosomes.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas Cromosómicas no Histona/genética , Cromosomas/genética , Proteínas Represoras/genética , Transcripción Genética , Factor de Unión a CCCTC , Regulación de la Expresión Génica , Genoma Humano , Humanos , Cohesinas
10.
Methods ; 142: 39-46, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29772275

RESUMEN

Mammalian genomes are folded in a hierarchy of compartments, topologically associating domains (TADs), subTADs, and looping interactions. Currently, there is a great need to evaluate the link between chromatin topology and genome function across many biological conditions and genetic perturbations. Hi-C can generate genome-wide maps of looping interactions but is intractable for high-throughput comparison of loops across multiple conditions due to the enormous number of reads (>6 Billion) required per library. Here, we describe 5C-ID, a new version of Chromosome-Conformation-Capture-Carbon-Copy (5C) with restriction digest and ligation performed in the nucleus (in situ Chromosome-Conformation-Capture (3C)) and ligation-mediated amplification performed with a double alternating primer design. We demonstrate that 5C-ID produces higher-resolution 3D genome folding maps with reduced spatial noise using markedly lower cell numbers than canonical 5C. 5C-ID enables the creation of high-resolution, high-coverage maps of chromatin loops in up to a 30 Megabase subset of the genome at a fraction of the cost of Hi-C.


Asunto(s)
Mapeo Cromosómico/métodos , Cromosomas/genética , Cartilla de ADN/genética , Genoma/genética , Conformación de Ácido Nucleico , Animales , Técnicas de Cultivo de Célula/métodos , Células Cultivadas , Cromosomas/química , Ratones , Ratones Endogámicos C57BL , Células Madre Embrionarias de Ratones , Reacción en Cadena de la Polimerasa , Análisis de Secuencia de ADN
11.
Genome Biol Evol ; 2024 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-39382451

RESUMEN

The 3D structure of the genome is an important mediator of gene expression. As phenotypic divergence is largely driven by gene regulatory variation, comparing genome 3D contacts across species can further understanding of the molecular basis of species differences. However, while experimental data on genome 3D contacts in humans is increasingly abundant, only a handful of 3D genome contact maps exist for other species. Here, we demonstrate that human experimental data can be used to close this data gap. We apply a machine learning model that predicts 3D genome contacts from DNA sequence to the genomes from 56 bonobos and chimpanzees and identify species-specific patterns of genome folding. We estimated 3D divergence between individuals from the resulting contact maps in 4,420 1 Mb genomic windows, of which ∼17% were substantially divergent in predicted genome contacts. Bonobos and chimpanzees diverged at 89 windows, overlapping genes associated with multiple traits implicated in Pan phenotypic divergence. We discovered 51 bonobo-specific variants that individually produce the observed bonobo contact pattern in bonobo-chimpanzee divergent windows. Our results demonstrate that machine learning methods can leverage human data to fill in data gaps across species, offering the first look at population-level 3D genome variation in non-human primates. We also identify loci where changes in 3D folding may contribute to phenotypic differences in our closest living relatives.

12.
Biology (Basel) ; 12(8)2023 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-37626945

RESUMEN

Three-dimensional genome organization has been increasingly recognized as an important determinant of the precise regulation of gene expression in mammalian cells, yet the relationship between gene transcriptional activity and spatial subcompartment positioning is still not fully comprehended. Here, we first utilized genome-wide Hi-C data to infer eight types of subcompartment (labeled A1, A2, A3, A4, B1, B2, B3, and B4) in mouse embryonic stem cells and four primary differentiated cell types, including thymocytes, macrophages, neural progenitor cells, and cortical neurons. Transitions of subcompartments may confer gene expression changes in different cell types. Intriguingly, we identified two subsets of subcompartments defined by higher gene density and characterized by strongly looped contact domains, named common A1 and variable A1, respectively. We revealed that common A1, which includes highly expressed genes and abundant housekeeping genes, shows a ~2-fold higher gene density than the variable A1, where cell type-specific genes are significantly enriched. Thus, our study supports a model in which both types of genomic loci with constitutive and regulatory high transcriptional activity can drive the subcompartment A1 formation. Special chromatin subcompartment arrangement and intradomain interactions may, in turn, contribute to maintaining proper levels of gene expression, especially for regulatory non-housekeeping genes.

13.
Cell Rep ; 42(4): 112361, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37059093

RESUMEN

Over the last decades, technological breakthroughs in super-resolution microscopy have allowed us to reach molecular resolution and design experiments of unprecedented complexity. Investigating how chromatin is folded in 3D, from the nucleosome level up to the entire genome, is becoming possible by "magic" (imaging genomic), i.e., the combination of imaging and genomic approaches. This offers endless opportunities to delve into the relationship between genome structure and function. Here, we review recently achieved objectives and the conceptual and technical challenges the field of genome architecture is currently undertaking. We discuss what we have learned so far and where we are heading. We elucidate how the different super-resolution microscopy approaches and, more specifically, live-cell imaging have contributed to the understanding of genome folding. Moreover, we discuss how future technical developments could address remaining open questions.


Asunto(s)
Cromatina , Nucleosomas , Cromatina/genética , Nucleosomas/genética , Genoma , Genómica
14.
Elife ; 112022 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-36579892

RESUMEN

The expression of a mutant Lamin A, progerin, in Hutchinson-Gilford Progeria Syndrome leads to alterations in genome architecture, nuclear morphology, epigenetic states, and altered phenotypes in all cells of the mesenchymal lineage. Here, we report a comprehensive analysis of the transcriptional status of patient derived HGPS fibroblasts, including nine cell lines not previously reported, in comparison with age-matched controls, adults, and old adults. We find that Progeria fibroblasts carry abnormal transcriptional signatures, centering around several functional hubs: DNA maintenance and epigenetics, bone development and homeostasis, blood vessel maturation and development, fat deposition and lipid management, and processes related to muscle growth. Stratification of patients by age revealed misregulated expression of genes related to endochondral ossification and chondrogenic commitment in children aged 4-7 years old, where this differentiation program starts in earnest. Hi-C measurements on patient fibroblasts show weakening of genome compartmentalization strength but increases in TAD strength. While the majority of gene misregulation occurs in regions which do not change spatial chromosome organization, some expression changes in key mesenchymal lineage genes coincide with lamin associated domain misregulation and shifts in genome compartmentalization.


Asunto(s)
Progeria , Humanos , Progeria/genética , Osteogénesis/genética , Diferenciación Celular , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Fibroblastos/metabolismo
15.
Comput Struct Biotechnol J ; 20: 3439-3448, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35832620

RESUMEN

Gene regulation in eukaryotes is profoundly shaped by the 3D organization of chromatin within the cell nucleus. Distal regulatory interactions between enhancers and their target genes are widespread and many causal loci underlying heritable agricultural or clinical traits have been mapped to distal cis-regulatory elements. Dissecting the sequence features that mediate such distal interactions is key to understanding their underlying biology. Deep Learning (DL) models coupled with genome-wide 3C-based sequencing data have emerged as powerful tools to infer the DNA sequence grammar underlying such distal interactions. In this review we show that most DL models have remarkably high prediction accuracy, which indicates that DNA sequence features are important determinants of chromatin looping. However, DL model training has so far been limited to a small set of human cell lines, raising questions about the generalization of these predictions to other tissue-types and species. Furthermore, we find that the model architecture seems less relevant for model performance than the training strategy and the data preparation step. Transfer learning, coupled with functionally curated interactions, appear to be the most promising approach to learn cell-type specific and possibly species- specific sequence features in future applications.

16.
Biology (Basel) ; 11(5)2022 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-35625436

RESUMEN

The cell nucleus is frequently considered a cage in which the genome is placed to protect it from various external factors. Inside the nucleus, many functional compartments have been identified that are directly or indirectly involved in implementing genomic DNA's genetic functions. For many years, it was assumed that these compartments are assembled on a proteinaceous scaffold (nuclear matrix), which provides a structural milieu for nuclear compartmentalization and genome folding while simultaneously offering some rigidity to the cell nucleus. The results of research in recent years have made it possible to consider the cell nucleus from a different angle. From the "box" in which the genome is placed, the nucleus has become a kind of mobile exoskeleton, which is formed around the packaged genome, under the influence of transcription and other processes directly related to the genome activity. In this review, we summarize the main arguments in favor of this point of view by analyzing the mechanisms that mediate cell nucleus assembly and support its resistance to mechanical stresses.

17.
J Mol Biol ; 432(3): 676-681, 2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-31689436

RESUMEN

How chromosomes are organized within the tridimensional space of the nucleus and how can this organization affect genome function have been long-standing questions on the path to understanding genome activity and its link to disease. In the last decade, high-throughput chromosome conformation capture techniques, such as Hi-C, have facilitated the discovery of new principles of genome folding. Chromosomes are folded in multiple high-order structures, with local contacts between enhancers and promoters, intermediate-level contacts forming Topologically Associating Domains (TADs) and higher-order chromatin structures sequestering chromatin into active and repressive compartments. However, despite the increasing evidence that genome organization can influence its function, we are still far from understanding the underlying mechanisms. Deciphering these mechanisms represents a major challenge for the future, which large, international initiatives, such as 4DN, HCA and LifeTime, aim to collaboratively tackle by using a conjunction of state-of-the-art population-based and single-cell approaches.


Asunto(s)
Cromatina/química , Cromatina/metabolismo , Regulación de la Expresión Génica , Genoma , Sustancias Macromoleculares/química , Sustancias Macromoleculares/metabolismo , Conformación Molecular , Animales , Investigación Biomédica/métodos , Investigación Biomédica/tendencias , Biología Molecular/métodos , Biología Molecular/tendencias
18.
Brief Funct Genomics ; 19(2): 71-82, 2020 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-31819944

RESUMEN

Chromosomes have a complex three-dimensional (3D) architecture comprising A/B compartments, topologically associating domains and promoter-enhancer interactions. At all these levels, the 3D genome has functional consequences for gene transcription and therefore for cellular identity. The development and activation of lymphocytes involves strict control of gene expression by transcription factors (TFs) operating in a three-dimensionally organized chromatin landscape. As lymphocytes are indispensable for tissue homeostasis and pathogen defense, and aberrant lymphocyte activity is involved in a wide range of human morbidities, acquiring an in-depth understanding of the molecular mechanisms that control lymphocyte identity is highly relevant. Here we review current knowledge of the interplay between 3D genome organization and transcriptional control during B and T lymphocyte development and antigen-dependent activation, placing special emphasis on the role of TFs.


Asunto(s)
Regiones Promotoras Genéticas/genética , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina/genética , Ensamble y Desensamble de Cromatina/fisiología , Humanos , Linfocitos/metabolismo
19.
Curr Top Dev Biol ; 133: 343-385, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30902258

RESUMEN

A complex cascade of highly regulated processes of cell fate determination, differentiation, proliferation and transdifferentiation dictate the patterning, morphogenesis and growth of the vertebrate skeleton, perturbation of which results in malformation. In humans over 450 different dysplasias involving the skeletal system constitute a significant fraction of documented Mendelian disorders. The combination of clinical, phenotypic characterization of rare human skeletal dysmorphologies, the discovery of causative mutations and functional validation in animal models has contributed enormously to the understanding of molecular control of skeletal development. These studies revealed a myriad of genes and pathways, such as WNT, Hedgehog (HH), planar cell polarity and primary cilia, as key regulators for skeletal patterning, growth and homeostasis. The generation of mouse models recapitulating human congenital skeletal dysplasia has provided mechanistic insights into the diverse pathologies caused by single gene mutations, integrated action of developmental pathways such as WNT and HH and the role of stress responses. Technological developments in whole genome and exome sequencing have accelerated the discovery of disease-causing mutations and are changing approaches for diagnosis. The discovery that non-coding variants and disorganization of the 3D genome are associated with limb patterning disorders has revealed an additional level of complexity in the regulatory framework of skeletal development and disease mechanisms. This chapter focuses on a selection of human skeletal pathologies which illustrate how new findings about the coding and noncoding genome, combined with functional modeling, are contributing to deeper understanding of skeletal development, mechanisms of disease, with therapeutic potential for chondrodysplasias.


Asunto(s)
Enfermedades Óseas/genética , Osteogénesis , Animales , Tipificación del Cuerpo/genética , Cilios/metabolismo , Estrés del Retículo Endoplásmico , Humanos , Osteogénesis/genética , Transducción de Señal
20.
Brief Funct Genomics ; 15(4): 305-14, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26206085

RESUMEN

Translating the vast amounts of genomic and epigenomic information accumulated on the linear genome into three-dimensional models of nuclear organization is a current major challenge. In response to this challenge, recent technological innovations based on chromosome conformation capture methods in combination with increasingly powerful functional approaches have revealed exciting insights into key aspects of genome regulation. These findings have led to an emerging model where the genome is folded and compartmentalized into highly conserved topological domains that are further divided into functional subdomains containing physical loops that bring cis-regulatory elements to close proximity. Targeted functional experiments, largely based on designable DNA-binding proteins, have begun to define the major architectural proteins required to establish and maintain appropriate genome regulation. Here, we focus on the accessible and well-characterized system of pluripotent cells to review the functional role of chromatin organization in regulating pluripotency, differentiation and reprogramming.


Asunto(s)
Diferenciación Celular , Ensamble y Desensamble de Cromatina/fisiología , Células Madre Pluripotentes/fisiología , Animales , Humanos , Células Madre Pluripotentes/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA