Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 4.683
Filtrar
Más filtros

Publication year range
1.
Annu Rev Biochem ; 92: 15-41, 2023 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-37137166

RESUMEN

SMC (structural maintenance of chromosomes) protein complexes are an evolutionarily conserved family of motor proteins that hold sister chromatids together and fold genomes throughout the cell cycle by DNA loop extrusion. These complexes play a key role in a variety of functions in the packaging and regulation of chromosomes, and they have been intensely studied in recent years. Despite their importance, the detailed molecular mechanism for DNA loop extrusion by SMC complexes remains unresolved. Here, we describe the roles of SMCs in chromosome biology and particularly review in vitro single-molecule studies that have recently advanced our understanding of SMC proteins. We describe the mechanistic biophysical aspects of loop extrusion that govern genome organization and its consequences.


Asunto(s)
Proteínas Cromosómicas no Histona , Complejos Multiproteicos , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Complejos Multiproteicos/química , Cromosomas/genética , Cromosomas/metabolismo , ADN/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo
2.
Cell ; 185(20): 3689-3704.e21, 2022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-36179666

RESUMEN

Regulatory landscapes drive complex developmental gene expression, but it remains unclear how their integrity is maintained when incorporating novel genes and functions during evolution. Here, we investigated how a placental mammal-specific gene, Zfp42, emerged in an ancient vertebrate topologically associated domain (TAD) without adopting or disrupting the conserved expression of its gene, Fat1. In ESCs, physical TAD partitioning separates Zfp42 and Fat1 with distinct local enhancers that drive their independent expression. This separation is driven by chromatin activity and not CTCF/cohesin. In contrast, in embryonic limbs, inactive Zfp42 shares Fat1's intact TAD without responding to active Fat1 enhancers. However, neither Fat1 enhancer-incompatibility nor nuclear envelope-attachment account for Zfp42's unresponsiveness. Rather, Zfp42's promoter is rendered inert to enhancers by context-dependent DNA methylation. Thus, diverse mechanisms enabled the integration of independent Zfp42 regulation in the Fat1 locus. Critically, such regulatory complexity appears common in evolution as, genome wide, most TADs contain multiple independently expressed genes.


Asunto(s)
Cromatina , Placenta , Animales , Factor de Unión a CCCTC/metabolismo , Ensamble y Desensamble de Cromatina , Elementos de Facilitación Genéticos , Evolución Molecular , Femenino , Genoma , Mamíferos/metabolismo , Placenta/metabolismo , Embarazo , Regiones Promotoras Genéticas , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
Cell ; 184(21): 5448-5464.e22, 2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34624221

RESUMEN

Structural maintenance of chromosomes (SMC) complexes organize genome topology in all kingdoms of life and have been proposed to perform this function by DNA loop extrusion. How this process works is unknown. Here, we have analyzed how loop extrusion is mediated by human cohesin-NIPBL complexes, which enable chromatin folding in interphase cells. We have identified DNA binding sites and large-scale conformational changes that are required for loop extrusion and have determined how these are coordinated. Our results suggest that DNA is translocated by a spontaneous 50 nm-swing of cohesin's hinge, which hands DNA over to the ATPase head of SMC3, where upon binding of ATP, DNA is clamped by NIPBL. During this process, NIPBL "jumps ship" from the hinge toward the SMC3 head and might thereby couple the spontaneous hinge swing to ATP-dependent DNA clamping. These results reveal mechanistic principles of how cohesin-NIPBL and possibly other SMC complexes mediate loop extrusion.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , ADN/química , Conformación de Ácido Nucleico , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Sitios de Unión , Proteínas de Ciclo Celular/química , ADN/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Células HeLa , Humanos , Hidrólisis , Cinética , Microscopía de Fuerza Atómica , Modelos Moleculares , Proteínas Nucleares/metabolismo , Conformación Proteica , Cohesinas
4.
Annu Rev Cell Dev Biol ; 38: 395-418, 2022 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-35850152

RESUMEN

Although tissue homeostasis-the steady state-implies stability, our organs are in a state of continual, large-scale cellular flux. This flux underpins an organ's ability to homeostatically renew, to non-homeostatically resize upon altered functional demand, and to return to homeostasis after resizing or injury-in other words, to be dynamic. Here, I examine the basic unit of organ-scale cell dynamics: the cellular life cycle of birth, differentiation, and death. Focusing on epithelial organs, I discuss how spatial patterns and temporal kinetics of life cycle stages depend upon lineage organization and tissue architecture. I review how signaling between stages coordinates life cycle dynamics to enforce homeostasis, and I highlight how particular stages are transiently unbalanced to drive organ resizing or repair. Finally, I offer that considering organs as a collective of not cells but rather cell life cycles provides a powerful vantage for deciphering homeostatic and non-homeostatic tissue states.


Asunto(s)
Transducción de Señal , Diferenciación Celular , Homeostasis
5.
Annu Rev Cell Dev Biol ; 37: 199-232, 2021 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-34228506

RESUMEN

Nuclei are central hubs for information processing in eukaryotic cells. The need to fit large genomes into small nuclei imposes severe restrictions on genome organization and the mechanisms that drive genome-wide regulatory processes. How a disordered polymer such as chromatin, which has vast heterogeneity in its DNA and histone modification profiles, folds into discernibly consistent patterns is a fundamental question in biology. Outstanding questions include how genomes are spatially and temporally organized to regulate cellular processes with high precision and whether genome organization is causally linked to transcription regulation. The advent of next-generation sequencing, super-resolution imaging, multiplexed fluorescent in situ hybridization, and single-molecule imaging in individual living cells has caused a resurgence in efforts to understand the spatiotemporal organization of the genome. In this review, we discuss structural and mechanistic properties of genome organization at different length scales and examine changes in higher-order chromatin organization during important developmental transitions.


Asunto(s)
Cromatina , Cromosomas , Cromatina/genética , ADN , Genoma , Hibridación Fluorescente in Situ
6.
Cell ; 173(6): 1508-1519.e18, 2018 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-29754816

RESUMEN

As predicted by the notion that sister chromatid cohesion is mediated by entrapment of sister DNAs inside cohesin rings, there is perfect correlation between co-entrapment of circular minichromosomes and sister chromatid cohesion. In most cells where cohesin loads without conferring cohesion, it does so by entrapment of individual DNAs. However, cohesin with a hinge domain whose positively charged lumen is neutralized loads and moves along chromatin despite failing to entrap DNAs. Thus, cohesin engages chromatin in non-topological, as well as topological, manners. Since hinge mutations, but not Smc-kleisin fusions, abolish entrapment, DNAs may enter cohesin rings through hinge opening. Mutation of three highly conserved lysine residues inside the Smc1 moiety of Smc1/3 hinges abolishes all loading without affecting cohesin's recruitment to CEN loading sites or its ability to hydrolyze ATP. We suggest that loading and translocation are mediated by conformational changes in cohesin's hinge driven by cycles of ATP hydrolysis.


Asunto(s)
Proteínas de Ciclo Celular/química , Cromátides/química , Proteínas Cromosómicas no Histona/química , ADN/química , Adenosina Trifosfato/química , Animales , Sitios de Unión , Cromatina/química , Humanos , Hidrólisis , Lisina/química , Ratones , Mutación , Proteínas Nucleares/genética , Conformación Proteica , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Cohesinas
7.
Cell ; 173(5): 1165-1178.e20, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29706548

RESUMEN

Cohesin extrusion is thought to play a central role in establishing the architecture of mammalian genomes. However, extrusion has not been visualized in vivo, and thus, its functional impact and energetics are unknown. Using ultra-deep Hi-C, we show that loop domains form by a process that requires cohesin ATPases. Once formed, however, loops and compartments are maintained for hours without energy input. Strikingly, without ATP, we observe the emergence of hundreds of CTCF-independent loops that link regulatory DNA. We also identify architectural "stripes," where a loop anchor interacts with entire domains at high frequency. Stripes often tether super-enhancers to cognate promoters, and in B cells, they facilitate Igh transcription and recombination. Stripe anchors represent major hotspots for topoisomerase-mediated lesions, which promote chromosomal translocations and cancer. In plasmacytomas, stripes can deregulate Igh-translocated oncogenes. We propose that higher organisms have coopted cohesin extrusion to enhance transcription and recombination, with implications for tumor development.


Asunto(s)
Adenosina Trifosfato/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Genoma , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Línea Celular , Proteoglicanos Tipo Condroitín Sulfato/genética , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/genética , Cromosomas/metabolismo , Proteínas de Unión al ADN , Humanos , Ratones , Mutagénesis , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Cohesinas
8.
Cell ; 174(1): 102-116.e14, 2018 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-29804837

RESUMEN

RAG endonuclease initiates antibody heavy chain variable region exon assembly from V, D, and J segments within a chromosomal V(D)J recombination center (RC) by cleaving between paired gene segments and flanking recombination signal sequences (RSSs). The IGCR1 control region promotes DJH intermediate formation by isolating Ds, JHs, and RCs from upstream VHs in a chromatin loop anchored by CTCF-binding elements (CBEs). How VHs access the DJHRC for VH to DJH rearrangement was unknown. We report that CBEs immediately downstream of frequently rearranged VH-RSSs increase recombination potential of their associated VH far beyond that provided by RSSs alone. This CBE activity becomes particularly striking upon IGCR1 inactivation, which allows RAG, likely via loop extrusion, to linearly scan chromatin far upstream. VH-associated CBEs stabilize interactions of D-proximal VHs first encountered by the DJHRC during linear RAG scanning and thereby promote dominant rearrangement of these VHs by an unanticipated chromatin accessibility-enhancing CBE function.


Asunto(s)
Factor de Unión a CCCTC/metabolismo , Cromatina/metabolismo , Proteínas de Homeodominio/metabolismo , Recombinación V(D)J , Animales , Línea Celular , ADN Intergénico/genética , ADN Intergénico/metabolismo , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/metabolismo , Región Variable de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Mutagénesis , Señales de Clasificación de Proteína , ARN Guía de Kinetoplastida/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo
9.
Annu Rev Biochem ; 86: 461-484, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28654322

RESUMEN

Self-catalyzed DNA depurination is a sequence-specific physiological mechanism mediated by spontaneous extrusion of a stem-loop catalytic intermediate. Hydrolysis of the 5'G residue of the 5'GA/TGG loop and of the first 5'A residue of the 5'GAGA loop, together with particular first stem base pairs, specifies their hydrolysis without involving protein, cofactor, or cation. As such, this mechanism is the only known DNA catalytic activity exploited by nature. The consensus sequences for self-depurination of such G- and A-loop residues occur in all genomes examined across the phyla, averaging one site every 2,000-4,000 base pairs. Because apurinic sites are subject to error-prone repair, leading to substitution and short frameshift mutations, they are both a source of genome damage and a means for creating sequence diversity. Their marked overrepresentation in genomes, and largely unchanging density from the lowest to the highest organisms, indicate their selection over the course of evolution. The mutagenicity at such sites in many human genes is associated with loss of function of key proteins responsible for diverse diseases.


Asunto(s)
Adenina/metabolismo , Síndrome de Bloom/genética , ADN Catalítico/genética , Guanina/metabolismo , Polimorfismo Genético , Síndrome de Werner/genética , Evolución Biológica , Síndrome de Bloom/metabolismo , Síndrome de Bloom/patología , Catálisis , Reparación del ADN , ADN Catalítico/metabolismo , ADN Cruciforme/genética , ADN Cruciforme/metabolismo , ADN de Cadena Simple/genética , ADN de Cadena Simple/metabolismo , Humanos , Hidrólisis , Secuencias Invertidas Repetidas , Mutación , Síndrome de Werner/metabolismo , Síndrome de Werner/patología , Globinas beta/genética , Globinas beta/metabolismo
10.
Cell ; 171(3): 588-600.e24, 2017 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-28988770

RESUMEN

Condensin protein complexes coordinate the formation of mitotic chromosomes and thereby ensure the successful segregation of replicated genomes. Insights into how condensin complexes bind to chromosomes and alter their topology are essential for understanding the molecular principles behind the large-scale chromatin rearrangements that take place during cell divisions. Here, we identify a direct DNA-binding site in the eukaryotic condensin complex, which is formed by its Ycg1Cnd3 HEAT-repeat and Brn1Cnd2 kleisin subunits. DNA co-crystal structures reveal a conserved, positively charged groove that accommodates the DNA double helix. A peptide loop of the kleisin subunit encircles the bound DNA and, like a safety belt, prevents its dissociation. Firm closure of the kleisin loop around DNA is essential for the association of condensin complexes with chromosomes and their DNA-stimulated ATPase activity. Our data suggest a sophisticated molecular basis for anchoring condensin complexes to chromosomes that enables the formation of large-sized chromatin loops.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Cromosomas/metabolismo , Proteínas de Unión al ADN/metabolismo , Eucariontes/metabolismo , Proteínas Fúngicas/metabolismo , Complejos Multiproteicos/metabolismo , Adenosina Trifosfatasas/química , Secuencia de Aminoácidos , Chaetomium/metabolismo , Cromosomas/química , Cristalografía por Rayos X , ADN/química , ADN/metabolismo , Proteínas de Unión al ADN/química , Eucariontes/química , Proteínas Fúngicas/química , Células HeLa , Humanos , Modelos Moleculares , Complejos Multiproteicos/química , Saccharomyces cerevisiae/metabolismo , Alineación de Secuencia
11.
Cell ; 171(2): 305-320.e24, 2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28985562

RESUMEN

The human genome folds to create thousands of intervals, called "contact domains," that exhibit enhanced contact frequency within themselves. "Loop domains" form because of tethering between two loci-almost always bound by CTCF and cohesin-lying on the same chromosome. "Compartment domains" form when genomic intervals with similar histone marks co-segregate. Here, we explore the effects of degrading cohesin. All loop domains are eliminated, but neither compartment domains nor histone marks are affected. Loss of loop domains does not lead to widespread ectopic gene activation but does affect a significant minority of active genes. In particular, cohesin loss causes superenhancers to co-localize, forming hundreds of links within and across chromosomes and affecting the regulation of nearby genes. We then restore cohesin and monitor the re-formation of each loop. Although re-formation rates vary greatly, many megabase-sized loops recovered in under an hour, consistent with a model where loop extrusion is rapid.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/genética , Proteínas Cromosómicas no Histona/metabolismo , Cromosomas/metabolismo , Genoma Humano , Proteínas Represoras/metabolismo , Factor de Unión a CCCTC , Línea Celular Tumoral , Proteínas de Unión al ADN , Elementos de Facilitación Genéticos , Código de Histonas , Humanos , Proteínas Nucleares/metabolismo , Nucleosomas/metabolismo , Fosfoproteínas/metabolismo , Cohesinas
12.
Cell ; 171(1): 103-119.e18, 2017 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-28938112

RESUMEN

It is now established that Bcl11b specifies T cell fate. Here, we show that in developing T cells, the Bcl11b enhancer repositioned from the lamina to the nuclear interior. Our search for factors that relocalized the Bcl11b enhancer identified a non-coding RNA named ThymoD (thymocyte differentiation factor). ThymoD-deficient mice displayed a block at the onset of T cell development and developed lymphoid malignancies. We found that ThymoD transcription promoted demethylation at CTCF bound sites and activated cohesin-dependent looping to reposition the Bcl11b enhancer from the lamina to the nuclear interior and to juxtapose the Bcl11b enhancer and promoter into a single-loop domain. These large-scale changes in nuclear architecture were associated with the deposition of activating epigenetic marks across the loop domain, plausibly facilitating phase separation. These data indicate how, during developmental progression and tumor suppression, non-coding transcription orchestrates chromatin folding and compartmentalization to direct with high precision enhancer-promoter communication.


Asunto(s)
Elementos de Facilitación Genéticos , Regiones Promotoras Genéticas , ARN no Traducido/genética , Proteínas Represoras/genética , Linfocitos T/citología , Proteínas Supresoras de Tumor/genética , Animales , Factor de Unión a CCCTC , Cromatina/metabolismo , Leucemia/genética , Región de Control de Posición , Linfoma/genética , Ratones , Lámina Nuclear/metabolismo , Proteínas Represoras/metabolismo , Linfocitos T/metabolismo , Timo/citología , Timo/metabolismo , Transcripción Genética
13.
Cell ; 169(4): 693-707.e14, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28475897

RESUMEN

The spatial organization of chromosomes influences many nuclear processes including gene expression. The cohesin complex shapes the 3D genome by looping together CTCF sites along chromosomes. We show here that chromatin loop size can be increased and that the duration with which cohesin embraces DNA determines the degree to which loops are enlarged. Cohesin's DNA release factor WAPL restricts this loop extension and also prevents looping between incorrectly oriented CTCF sites. We reveal that the SCC2/SCC4 complex promotes the extension of chromatin loops and the formation of topologically associated domains (TADs). Our data support the model that cohesin structures chromosomes through the processive enlargement of loops and that TADs reflect polyclonal collections of loops in the making. Finally, we find that whereas cohesin promotes chromosomal looping, it rather limits nuclear compartmentalization. We conclude that the balanced activity of SCC2/SCC4 and WAPL enables cohesin to correctly structure chromosomes.


Asunto(s)
Proteínas Portadoras/metabolismo , Núcleo Celular/metabolismo , Cromatina/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Acetiltransferasas/metabolismo , Factor de Unión a CCCTC , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN , Elongasas de Ácidos Grasos , Edición Génica , Humanos , Complejos Multiproteicos/metabolismo , Proteínas Represoras/metabolismo , Cohesinas
14.
Annu Rev Cell Dev Biol ; 34: 311-332, 2018 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-30089222

RESUMEN

Balancing cell death and survival is essential for normal development and homeostasis and for preventing diseases, especially cancer. Conventional cell death pathways include apoptosis, a form of programmed cell death controlled by a well-defined biochemical pathway, and necrosis, the lysis of acutely injured cells. New types of regulated cell death include necroptosis, pyroptosis, ferroptosis, phagoptosis, and entosis. Autophagy can promote survival or can cause death. Newly described processes of anastasis and resuscitation show that, remarkably, cells can recover from the brink of apoptosis or necroptosis. Important new work shows that epithelia achieve homeostasis by extruding excess cells, which then die by anoikis due to loss of survival signals. This mechanically regulated process both maintains barrier function as cells die and matches rates of proliferation and death. In this review, we describe these unconventional ways in which cells have evolved to die or survive, as well as the contributions that these processes make to homeostasis and cancer.


Asunto(s)
Apoptosis/genética , Autofagia/genética , Necrosis/genética , Neoplasias/genética , Anoicis/genética , Proliferación Celular/genética , Entosis/genética , Homeostasis/genética , Humanos , Piroptosis/genética , Transducción de Señal/genética
15.
Mol Cell ; 84(6): 1139-1148.e5, 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38452765

RESUMEN

Eukaryotic genomes are folded into DNA loops mediated by structural maintenance of chromosomes (SMC) complexes such as cohesin, condensin, and Smc5/6. This organization regulates different DNA-related processes along the cell cycle, such as transcription, recombination, segregation, and DNA repair. During the G2 stage, SMC-mediated DNA loops coexist with cohesin complexes involved in sister chromatid cohesion (SCC). However, the articulation between the establishment of SCC and the formation of SMC-mediated DNA loops along the chromatin remains unknown. Here, we show that SCC is indeed a barrier to cohesin-mediated DNA loop expansion along G2/M Saccharomyces cerevisiae chromosomes.


Asunto(s)
Proteínas Cromosómicas no Histona , Proteínas de Saccharomyces cerevisiae , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cromátides/genética , Cromátides/metabolismo , Cohesinas , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , ADN/genética , ADN/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
16.
Mol Cell ; 84(5): 867-882.e5, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38295804

RESUMEN

The structural maintenance of chromosomes (SMC) protein complexes-cohesin, condensin, and the Smc5/6 complex (Smc5/6)-are essential for chromosome function. At the molecular level, these complexes fold DNA by loop extrusion. Accordingly, cohesin creates chromosome loops in interphase, and condensin compacts mitotic chromosomes. However, the role of Smc5/6's recently discovered DNA loop extrusion activity is unknown. Here, we uncover that Smc5/6 associates with transcription-induced positively supercoiled DNA at cohesin-dependent loop boundaries on budding yeast (Saccharomyces cerevisiae) chromosomes. Mechanistically, single-molecule imaging reveals that dimers of Smc5/6 specifically recognize the tip of positively supercoiled DNA plectonemes and efficiently initiate loop extrusion to gather the supercoiled DNA into a large plectonemic loop. Finally, Hi-C analysis shows that Smc5/6 links chromosomal regions containing transcription-induced positive supercoiling in cis. Altogether, our findings indicate that Smc5/6 controls the three-dimensional organization of chromosomes by recognizing and initiating loop extrusion on positively supercoiled DNA.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas de Saccharomyces cerevisiae , Proteínas de Ciclo Celular/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , ADN Superhelicoidal/genética , Cohesinas , ADN/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Cromosomas/metabolismo
17.
Mol Cell ; 84(5): 883-896.e7, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38309275

RESUMEN

DNA loop-extruding SMC complexes play crucial roles in chromosome folding and DNA immunity. Prokaryotic SMC Wadjet (JET) complexes limit the spread of plasmids through DNA cleavage, yet the mechanisms for plasmid recognition are unresolved. We show that artificial DNA circularization renders linear DNA susceptible to JET nuclease cleavage. Unlike free DNA, JET cleaves immobilized plasmid DNA at a specific site, the plasmid-anchoring point, showing that the anchor hinders DNA extrusion but not DNA cleavage. Structures of plasmid-bound JetABC reveal two presumably stalled SMC motor units that are drastically rearranged from the resting state, together entrapping a U-shaped DNA segment, which is further converted to kinked V-shaped cleavage substrate by JetD nuclease binding. Our findings uncover mechanical bending of residual unextruded DNA as molecular signature for plasmid recognition and non-self DNA elimination. We moreover elucidate key elements of SMC loop extrusion, including the motor direction and the structure of a DNA-holding state.


Asunto(s)
ADN , Endonucleasas , ADN/metabolismo , Plásmidos/genética , Células Procariotas , Proteínas de Ciclo Celular/metabolismo
18.
Mol Cell ; 83(17): 3049-3063.e6, 2023 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-37591243

RESUMEN

Cohesin connects CTCF-binding sites and other genomic loci in cis to form chromatin loops and replicated DNA molecules in trans to mediate sister chromatid cohesion. Whether cohesin uses distinct or related mechanisms to perform these functions is unknown. Here, we describe a cohesin hinge mutant that can extrude DNA into loops but is unable to mediate cohesion in human cells. Our results suggest that the latter defect arises during cohesion establishment. The observation that cohesin's cohesion and loop extrusion activities can be partially separated indicates that cohesin uses distinct mechanisms to perform these two functions. Unexpectedly, the same hinge mutant can also not be stopped by CTCF boundaries as well as wild-type cohesin. This suggests that cohesion establishment and cohesin's interaction with CTCF boundaries depend on related mechanisms and raises the possibility that both require transient hinge opening to entrap DNA inside the cohesin ring.


Asunto(s)
Proteínas de Ciclo Celular , Cromátides , Humanos , Cromátides/genética , Sitios de Unión , Proteínas de Ciclo Celular/genética , Proteínas Cromosómicas no Histona/genética , Cohesinas
19.
Mol Cell ; 83(5): 681-697.e7, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36736317

RESUMEN

Interactions between transcription and cohesin-mediated loop extrusion can influence 3D chromatin architecture. However, their relevance in biology is unclear. Here, we report a direct role for such interactions in the mechanism of antibody class switch recombination (CSR) at the murine immunoglobulin heavy chain locus (Igh). Using Tri-C to measure higher-order multiway interactions on single alleles, we find that the juxtaposition (synapsis) of transcriptionally active donor and acceptor Igh switch (S) sequences, an essential step in CSR, occurs via the interaction of loop extrusion complexes with a de novo topologically associating domain (TAD) boundary formed via transcriptional activity across S regions. Surprisingly, synapsis occurs predominantly in proximity to the 3' CTCF-binding element (3'CBE) rather than the Igh super-enhancer, suggesting a two-step mechanism whereby transcription of S regions is not topologically coupled to synapsis, as has been previously proposed. Altogether, these insights advance our understanding of how 3D chromatin architecture regulates CSR.


Asunto(s)
Reordenamiento Génico , Cadenas Pesadas de Inmunoglobulina , Ratones , Animales , Cadenas Pesadas de Inmunoglobulina/genética , Cambio de Clase de Inmunoglobulina , Cromatina , Isotipos de Inmunoglobulinas
20.
Mol Cell ; 83(9): 1446-1461.e6, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-36996812

RESUMEN

Enhancer clusters overlapping disease-associated mutations in Pierre Robin sequence (PRS) patients regulate SOX9 expression at genomic distances over 1.25 Mb. We applied optical reconstruction of chromatin architecture (ORCA) imaging to trace 3D locus topology during PRS-enhancer activation. We observed pronounced changes in locus topology between cell types. Subsequent analysis of single-chromatin fiber traces revealed that these ensemble-average differences arise through changes in the frequency of commonly sampled topologies. We further identified two CTCF-bound elements, internal to the SOX9 topologically associating domain, which promote stripe formation, are positioned near the domain's 3D geometric center, and bridge enhancer-promoter contacts in a series of chromatin loops. Ablation of these elements results in diminished SOX9 expression and altered domain-wide contacts. Polymer models with uniform loading across the domain and frequent cohesin collisions recapitulate this multi-loop, centrally clustered geometry. Together, we provide mechanistic insights into architectural stripe formation and gene regulation over ultra-long genomic ranges.


Asunto(s)
Cromatina , Secuencias Reguladoras de Ácidos Nucleicos , Humanos , Cromatina/genética , Regiones Promotoras Genéticas , Regulación de la Expresión Génica , Genoma , Proteínas de Ciclo Celular/metabolismo , Elementos de Facilitación Genéticos , Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda