Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Cell Biol ; 223(6)2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38767621

RESUMEN

In this issue, the discovery by Yang et al. (https://doi.org/10.1083/jcb.202308013) that folded WW domains of YAP1 and other proteins bind to Impα introduces a new class of globular NLS, contrasting with the extensively studied linear NLS motifs. This finding underscores the versatility of importins in recognizing their cargo proteins.


Asunto(s)
Señales de Localización Nuclear , Humanos , Señales de Localización Nuclear/metabolismo , Dominios WW/genética , alfa Carioferinas/metabolismo , alfa Carioferinas/genética , alfa Carioferinas/química , Unión Proteica , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/química , Proteínas Señalizadoras YAP/metabolismo
3.
Proc Natl Acad Sci U S A ; 120(29): e2301199120, 2023 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-37450495

RESUMEN

Previously, we showed that the nuclear import receptor Importin-9 wraps around the H2A-H2B core to chaperone and transport it from the cytoplasm to the nucleus. However, unlike most nuclear import systems where RanGTP dissociates cargoes from their importins, RanGTP binds stably to the Importin-9•H2A-H2B complex, and formation of the ternary RanGTP•Importin-9•H2A-H2B complex facilitates H2A-H2B release to the assembling nucleosome. It was unclear how RanGTP and the cargo H2A-H2B can bind simultaneously to an importin, and how interactions of the three components position H2A-H2B for release. Here, we show cryo-EM structures of Importin-9•RanGTP and of its yeast homolog Kap114, including Kap114•RanGTP, Kap114•H2A-H2B, and RanGTP•Kap114•H2A-H2B, to explain how the conserved Kap114 binds H2A-H2B and RanGTP simultaneously and how the GTPase primes histone transfer to the nucleosome. In the ternary complex, RanGTP binds to the N-terminal repeats of Kap114 in the same manner as in the Kap114/Importin-9•RanGTP complex, and H2A-H2B binds via its acidic patch to the Kap114 C-terminal repeats much like in the Kap114/Importin-9•H2A-H2B complex. Ran binds to a different conformation of Kap114 in the ternary RanGTP•Kap114•H2A-H2B complex. Here, Kap114 no longer contacts the H2A-H2B surface proximal to the H2A docking domain that drives nucleosome assembly, positioning it for transfer to the assembling nucleosome or to dedicated H2A-H2B chaperones in the nucleus.


Asunto(s)
Nucleosomas , Proteínas de Saccharomyces cerevisiae , Nucleosomas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Unión Proteica , Carioferinas/metabolismo , Saccharomyces cerevisiae/metabolismo , Chaperonas Moleculares/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo
4.
J Clin Invest ; 133(14)2023 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-37463454

RESUMEN

Mutations in HNRNPH2 cause an X-linked neurodevelopmental disorder with features that include developmental delay, motor function deficits, and seizures. More than 90% of patients with hnRNPH2 have a missense mutation within or adjacent to the nuclear localization signal (NLS) of hnRNPH2. Here, we report that hnRNPH2 NLS mutations caused reduced interaction with the nuclear transport receptor Kapß2 and resulted in modest cytoplasmic accumulation of hnRNPH2. We generated 2 knockin mouse models with human-equivalent mutations in Hnrnph2 as well as Hnrnph2-KO mice. Knockin mice recapitulated clinical features of the human disorder, including reduced survival in male mice, impaired motor and cognitive functions, and increased susceptibility to audiogenic seizures. In contrast, 2 independent lines of Hnrnph2-KO mice showed no detectable phenotypes. Notably, KO mice had upregulated expression of Hnrnph1, a paralog of Hnrnph2, whereas knockin mice failed to upregulate Hnrnph1. Thus, genetic compensation by Hnrnph1 may counteract the loss of hnRNPH2. These findings suggest that HNRNPH2-related disorder may be driven by a toxic gain of function or a complex loss of HNRNPH2 function with impaired compensation by HNRNPH1. The knockin mice described here are an important resource for preclinical studies to assess the therapeutic benefit of gene replacement or knockdown of mutant hnRNPH2.


Asunto(s)
Trastornos del Neurodesarrollo , Animales , Humanos , Masculino , Ratones , Modelos Animales de Enfermedad , Mutación , Mutación Missense , Convulsiones/genética
5.
Structure ; 31(8): 924-934.e4, 2023 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-37279758

RESUMEN

The HNRNPH2 proline-tyrosine nuclear localization signal (PY-NLS) is mutated in HNRNPH2-related X-linked neurodevelopmental disorder, causing the normally nuclear HNRNPH2 to accumulate in the cytoplasm. We solved the cryoelectron microscopy (cryo-EM) structure of Karyopherin-ß2/Transportin-1 bound to the HNRNPH2 PY-NLS to understand importin-NLS recognition and disruption in disease. HNRNPH2 206RPGPY210 is a typical R-X2-4-P-Y motif comprising PY-NLS epitopes 2 and 3, followed by an additional Karyopherin-ß2-binding epitope, we term epitope 4, at residues 211DRP213; no density is present for PY-NLS epitope 1. Disease variant mutations at epitopes 2-4 impair Karyopherin-ß2 binding and cause aberrant cytoplasmic accumulation in cells, emphasizing the role of nuclear import defect in disease. Sequence/structure analysis suggests that strong PY-NLS epitopes 4 are rare and thus far limited to close paralogs of HNRNPH2, HNRNPH1, and HNRNPF. Epitope 4-binidng hotspot Karyopherin-ß2 W373 corresponds to close paralog Karyopherin-ß2b/Transportin-2 W370, a pathological variant site in neurodevelopmental abnormalities, suggesting that Karyopherin-ß2b/Transportin-2-HNRNPH2/H1/F interactions may be compromised in the abnormalities.


Asunto(s)
Carioferinas , Señales de Localización Nuclear , Carioferinas/metabolismo , Señales de Localización Nuclear/metabolismo , Epítopos/metabolismo , Tirosina/metabolismo , Prolina , Microscopía por Crioelectrón , Transporte Activo de Núcleo Celular , beta Carioferinas/genética , beta Carioferinas/química , beta Carioferinas/metabolismo , Núcleo Celular/metabolismo
6.
Structure ; 31(8): 903-911.e3, 2023 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-37379840

RESUMEN

Imp9 is the primary importin for shuttling H2A-H2B from the cytoplasm to the nucleus. It employs an unusual mechanism where the binding of RanGTP is insufficient to release H2A-H2B. The resulting stable RanGTP·Imp9·H2A-H2B complex gains nucleosome assembly activity with H2A-H2B able to be deposited into an assembling nucleosome in vitro. Using hydrogen-deuterium exchange coupled with mass spectrometry (HDX), we show that Imp9 stabilizes H2A-H2B beyond the direct-binding site, like other histone chaperones. HDX also shows that binding of RanGTP releases H2A-H2B contacts at Imp9 HEAT repeats 4-5, but not 18-19. DNA- and histone-binding surfaces of H2A-H2B are exposed in the ternary complex, facilitating nucleosome assembly. We also reveal that RanGTP has a weaker affinity for Imp9 when H2A-H2B is bound. Imp9 thus provides a connection between the nuclear import of H2A-H2B and its deposition into chromatin.


Asunto(s)
Histonas , Nucleosomas , Histonas/metabolismo , Proteína 1 de Ensamblaje de Nucleosomas/genética , Cromatina , Carioferinas/metabolismo
7.
bioRxiv ; 2023 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-37214964

RESUMEN

Histones are synthesized and processed in the cytoplasm and then transported into the nucleus for assembly into nucleosomes. H2A-H2B is imported into the S. cerevisiae nucleus by the importin Kap114, which also imports the most prominent H2A-H2B chaperone, Nap1. We understand how Kap114 recognizes H2A-H2B for nuclear import, but little is known about how it recognizes Nap1. Furthermore, the ternary complex of Nap1, H2A-H2B and Kap114 was previously detected in both the cytosol and the nucleus, but its role in nuclear import is unclear. Here, we present biophysical analysis of interactions between Nap1, H2A-H2B, Kap114 and RanGTP, and cryo-electron microscopy structures of ternary Kap114, Nap1 and H2A-H2B complexes. Kap114 binds Nap1 very weakly, but H2A-H2B enhances Kap114-Nap1 interaction to form a ternary Kap114/Nap1/H2A-H2B complex that is stable in the absence and presence of RanGTP. Cryogenic electron microscopy structures reveal two distinct ternary Kap114/Nap1/H2A-H2B complexes: a 3.2 Šresolution structure of Nap1 bound to H2A-H2B-bound Kap114 where Nap1 does not contact H2A-H2B, and a 3.5 Šresolution structure of H2A-H2B sandwiched between Nap1 and Kap114. Collectively, these results lead to a mechanistic model of how Nap1•H2A-H2B encounters Kap114 in the cytoplasm and how both H2A-H2B and Nap1 are chaperoned and co-imported by Kap114 into the nucleus. The model also suggests how RanGTP-binding stabilizes a quaternary RanGTP/Kap114/Nap1/H2A-H2B complex that facilitates hand-off of H2A-H2B from Kap114 to Nap1, the assembling nucleosome or other nuclear chaperone. Significance Statement: Free core histones are highly toxic and must be sequestered by other macromolecules in the cell. The mechanism of H3-H4 import by karyopherin Importin-4 in the presence of its chaperone ASF1 is understood, but the mechanism of how histone chaperone Nap1 influences H2A-H2B import is not resolved. We present biophysical interaction analysis and cryo-EM structures that reveal how Kap114, Nap1 and H2A-H2B assemble into an import complex. These results lead us to a structural mechanism of how Nap1 delivers H2A-H2B to Kap114 in the cytosol, how Nap1 and H2A-H2B are co-imported into the nucleus, and how RanGTP may influence Kap114/Nap1/H2A-H2B interactions to assemble nucleosomes in the nucleus.

8.
bioRxiv ; 2023 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-36747879

RESUMEN

Padavannil et al. 2019 show that Importin-9 (Imp9) transports Histones H2A-H2B from the cytoplasm to the nucleus using a non-canonical mechanism whereby binding of a GTP-bound Ran GTPase (RanGTP) fails to evict the H2A-H2B cargo. Instead, a stable complex forms, comprised of equimolar RanGTP, Imp9, and H2A-H2B. Unlike the binary Imp9•H2A-H2B complex, this RanGTP•Imp9•H2A-H2B ternary complex can release H2A-H2B to an assembling nucleosome. Here, we define the molecular basis for this RanGTP-activated nucleosome assembly by Imp9. We use hydrogen-deuterium exchange coupled with mass spectrometry and compare the dynamics and interfaces of the RanGTP•Imp9•H2A-H2B ternary complex to those in the Imp9•H2A-H2B or Imp9•RanGTP binary complexes. Our data are consistent with the Imp9•H2A-H2B structure by Padavannil et al. 2019 showing that Imp9 HEAT repeats 4-5 and 18-19 contact H2A-H2B, as well as many homologous importin•RanGTP structures showing that importin HEAT repeats 1 and 3, and the h8 loop, contact RanGTP. We show that Imp9 stabilizes H2A-H2B beyond the direct binding site, similar to other histone chaperones. Importantly, we reveal that binding of RanGTP releases H2A-H2B interaction at Imp9 HEAT repeats 4-5, but not 18-19. This exposes DNA- and histone-binding surfaces of H2A-H2B, thereby facilitating nucleosome assembly. We also reveal that RanGTP has a weaker affinity for Imp9 when H2A-H2B is bound. This may ensure that H2A-H2B is only released in high RanGTP concentrations near chromatin. We delineate the molecular link between the nuclear import of H2A-H2B and its deposition into chromatin by Imp9. Significance: Imp9 is the primary importin for shuttling H2A-H2B from the cytoplasm to the nucleus. It employs an unusual mechanism where the binding of RanGTP alone is insufficient to release H2A-H2B. The resulting stable RanGTP•Imp9•H2A-H2B complex gains nucleosome assembly activity as H2A-H2B can be deposited onto an assembling nucleosome. We show that H2A-H2B is allosterically stabilized via interactions with both N- and C-terminal portions of Imp9, reinforcing its chaperone-like behavior. RanGTP binding causes H2A-H2B release from the N-terminal portion of Imp9 only. The newly-exposed H2A-H2B surfaces can interact with DNA or H3-H4 in nucleosome assembly. Imp9 thus plays a multi-faceted role in histone import, storage, and deposition regulated by RanGTP, controlling histone supply in the nucleus and to chromatin.

9.
bioRxiv ; 2023 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-36711837

RESUMEN

The normally nuclear HNRNPH2 is mutated in HNRNPH2 -related X-linked neurodevelopmental disorder causing the protein to accumulate in the cytoplasm. Interactions of HNRNPH2 with its importin Karyopherin-ß2 (Transportin-1) had not been studied. We present a structure that shows Karyopherin-ß2 binding HNRNPH2 residues 204-215, a proline-tyrosine nuclear localization signal or PY-NLS that contains a typical R-X 2-4 -P-Y motif, 206 RPGPY 210 , followed a new Karyopherin-ß2 binding epitope at 211 DRP 213 that make many interactions with Karyopherin-ß2 W373. Mutations at each of these sites decrease Karyopherin-ß2 binding affinities by 70-100 fold, explaining aberrant accumulation in cells and emphasizing the role of nuclear import defects in the disease. Sequence/structure analysis suggests that the new epitope C-terminal of the PY-motif, which binds Karyopherin-ß2 W373, is rare and thus far limited to close paralogs HNRNPH2, HNRNPH1 and HNRNPF. Karyopherin-ß2 W373, a HNRNPH2-binding hotspot, corresponds to W370 of close paralog Transportin-2, a site of pathological variants in patients with neurodevelopmental abnormalities, suggesting that Transportin-2-HNRNPH2/H1/F interactions may be compromised in the abnormalities. Summary: HNRNPH2 variants in HNRNPH2 -related X-linked neurodevelopmental disorder aberrantly accumulate in the cytoplasm. A structure of Karyopherin-ß2•HNRNPH2 explains nuclear import defects of the variants, reveals a new NLS epitope that suggests mechanistic changes in pathological variants of Karyopherin-ß2 paralog Transportin-2.

10.
Proc Natl Acad Sci U S A ; 119(38): e2207177119, 2022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-36103578

RESUMEN

IMPORTIN-4, the primary nuclear import receptor of core histones H3 and H4, binds the H3-H4 dimer and histone chaperone ASF1 prior to nuclear import. However, how H3-H3-ASF1 is recognized for transport cannot be explained by available crystal structures of IMPORTIN-4-histone tail peptide complexes. Our 3.5-Å IMPORTIN-4-H3-H4-ASF1 cryoelectron microscopy structure reveals the full nuclear import complex and shows a binding mode different from suggested by previous structures. The N-terminal half of IMPORTIN-4 clamps the globular H3-H4 domain and H3 αN helix, while its C-terminal half binds the H3 N-terminal tail weakly; tail contribution to binding energy is negligible. ASF1 binds H3-H4 without contacting IMPORTIN-4. Together, ASF1 and IMPORTIN-4 shield nucleosomal H3-H4 surfaces to chaperone and import it into the nucleus where RanGTP binds IMPORTIN-4, causing large conformational changes to release H3-H4-ASF1. This work explains how full-length H3-H4 binds IMPORTIN-4 in the cytoplasm and how it is released in the nucleus.


Asunto(s)
Chaperonas de Histonas , Histonas , Carioferinas , Proteínas de Transporte de Membrana , Chaperonas Moleculares , Proteínas de Saccharomyces cerevisiae , Núcleo Celular/metabolismo , Microscopía por Crioelectrón , Citoplasma/metabolismo , Chaperonas de Histonas/química , Histonas/química , Humanos , Carioferinas/química , Proteínas de Transporte de Membrana/química , Chaperonas Moleculares/química , Conformación Proteica , Multimerización de Proteína , Proteínas de Saccharomyces cerevisiae/química
11.
Methods Mol Biol ; 2502: 245-256, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35412243

RESUMEN

CRM1 recognizes hundreds to thousands of protein cargoes by binding to the eight to fifteen residue-long nuclear export signals (NESs) within their polypeptide chains. Various assays to measure the binding affinity of NESs for CRM1 have been developed. CRM1 binds to NESs with a wide range of binding affinities, with dissociation constants that span from low nanomolar to tens of micromolar. An optimized binding affinity assay with improved throughput was recently developed to measure binding affinities of NES peptides for CRM1 in the presence of excess RanGTP. The assay can measure affinities, with multiple replicates, for up to seven different NES peptides per screening plate. Here, we present a protocol for the purification of the necessary proteins and for measuring CRM1-NES binding affinities.


Asunto(s)
Carioferinas , Señales de Exportación Nuclear , Receptores Citoplasmáticos y Nucleares , Transporte Activo de Núcleo Celular , Núcleo Celular/metabolismo , Carioferinas/química , Carioferinas/metabolismo , Péptidos/metabolismo , Unión Proteica , Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteína Exportina 1
12.
Methods Mol Biol ; 2502: 285-297, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35412246

RESUMEN

The Karyopherin protein CRM1 or XPO1 is the major nuclear export receptor that regulates nuclear exit of thousands of macromolecules in the cell. CRM1 recognizes protein cargoes by binding to their 8-15 residue-long nuclear export signals (NESs). A ternary CRM1-Ran-RanBP1 complex engineered to be suitable for crystallization has enabled structure determination by X-ray crystallography of CRM1 bound to many NES peptides and small-molecule inhibitors. Here, we present a protocol for the purification of the individual proteins, formation of the ternary CRM1-Ran-RanBP1 complex and crystallization of this complex for X-ray crystallography.


Asunto(s)
Cristalografía por Rayos X , Carioferinas , Señales de Exportación Nuclear , Receptores Citoplasmáticos y Nucleares , Transporte Activo de Núcleo Celular , Núcleo Celular/metabolismo , Cristalización/métodos , Cristalografía por Rayos X/métodos , Carioferinas/química , Carioferinas/metabolismo , Unión Proteica , Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteína Exportina 1
13.
Nat Rev Mol Cell Biol ; 23(5): 307-328, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35058649

RESUMEN

Efficient and regulated nucleocytoplasmic trafficking of macromolecules to the correct subcellular compartment is critical for proper functions of the eukaryotic cell. The majority of the macromolecular traffic across the nuclear pores is mediated by the Karyopherin-ß (or Kap) family of nuclear transport receptors. Work over more than two decades has shed considerable light on how the different Kap family members bring their respective cargoes into the nucleus or the cytoplasm in efficient and highly regulated manners. In this Review, we overview the main features and established functions of Kap family members, describe how Kaps recognize their cargoes and discuss the different ways in which these Kap-cargo interactions can be regulated, highlighting new findings and open questions. We also describe current knowledge of the import and export of the components of three large gene expression machines - the core replisome, RNA polymerase II and the ribosome - pointing out the questions that persist about how such large macromolecular complexes are trafficked to serve their function in a designated subcellular location.


Asunto(s)
Carioferinas , beta Carioferinas , Transporte Activo de Núcleo Celular/fisiología , Núcleo Celular/metabolismo , Carioferinas/metabolismo , Poro Nuclear/metabolismo , Receptores Citoplasmáticos y Nucleares , beta Carioferinas/metabolismo
15.
Sci Rep ; 11(1): 3754, 2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33580145

RESUMEN

Mutations in the RNA-binding protein FUS cause familial amyotropic lateral sclerosis (ALS). Several mutations that affect the proline-tyrosine nuclear localization signal (PY-NLS) of FUS cause severe juvenile ALS. FUS also undergoes liquid-liquid phase separation (LLPS) to accumulate in stress granules when cells are stressed. In unstressed cells, wild type FUS resides predominantly in the nucleus as it is imported by the importin Karyopherin-ß2 (Kapß2), which binds with high affinity to the C-terminal PY-NLS of FUS. Here, we analyze the interactions between two ALS-related variants FUS(P525L) and FUS(R495X) with importins, especially Kapß2, since they are still partially localized to the nucleus despite their defective/missing PY-NLSs. The crystal structure of the Kapß2·FUS(P525L)PY-NLS complex shows the mutant peptide making fewer contacts at the mutation site, explaining decreased affinity for Kapß2. Biochemical analysis revealed that the truncated FUS(R495X) protein, although missing the PY-NLS, can still bind Kapß2 and suppresses LLPS. FUS(R495X) uses its C-terminal tandem arginine-glycine-glycine regions, RGG2 and RGG3, to bind the PY-NLS binding site of Kapß2 for nuclear localization in cells when arginine methylation is inhibited. These findings suggest the importance of the C-terminal RGG regions in nuclear import and LLPS regulation of ALS variants of FUS that carry defective PY-NLSs.


Asunto(s)
Proteína FUS de Unión a ARN/metabolismo , beta Carioferinas/metabolismo , Transporte Activo de Núcleo Celular , Esclerosis Amiotrófica Lateral/genética , Sitios de Unión , Núcleo Celular/metabolismo , Humanos , Carioferinas/genética , Carioferinas/metabolismo , Señales de Localización Nuclear/genética , Unión Proteica , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/ultraestructura , beta Carioferinas/genética , beta Carioferinas/ultraestructura
16.
J Hematol Oncol ; 14(1): 17, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33451349

RESUMEN

BACKGROUND: Exportin 1 (XPO1/CRM1) is a key mediator of nuclear export with relevance to multiple cancers, including chronic lymphocytic leukemia (CLL). Whole exome sequencing has identified hot-spot somatic XPO1 point mutations which we found to disrupt highly conserved biophysical interactions in the NES-binding groove, conferring novel cargo-binding abilities and forcing cellular mis-localization of critical regulators. However, the pathogenic role played by change-in-function XPO1 mutations in CLL is not fully understood. METHODS: We performed a large, multi-center retrospective analysis of CLL cases (N = 1286) to correlate nonsynonymous mutations in XPO1 (predominantly E571K or E571G; n = 72) with genetic and epigenetic features contributing to the overall outcomes in these patients. We then established a mouse model with over-expression of wildtype (wt) or mutant (E571K or E571G) XPO1 restricted to the B cell compartment (Eµ-XPO1). Eµ-XPO1 mice were then crossed with the Eµ-TCL1 CLL mouse model. Lastly, we determined crystal structures of XPO1 (wt or E571K) bound to several selective inhibitors of nuclear export (SINE) molecules (KPT-185, KPT-330/Selinexor, and KPT-8602/Eltanexor). RESULTS: We report that nonsynonymous mutations in XPO1 associate with high risk genetic and epigenetic features and accelerated CLL progression. Using the newly-generated Eµ-XPO1 mouse model, we found that constitutive B-cell over-expression of wt or mutant XPO1 could affect development of a CLL-like disease in aged mice. Furthermore, concurrent B-cell expression of XPO1 with E571K or E571G mutations and TCL1 accelerated the rate of leukemogenesis relative to that of Eµ-TCL1 mice. Lastly, crystal structures of E571 or E571K-XPO1 bound to SINEs, including Selinexor, are highly similar, suggesting that the activity of this class of compounds will not be affected by XPO1 mutations at E571 in patients with CLL. CONCLUSIONS: These findings indicate that mutations in XPO1 at E571 can drive leukemogenesis by priming the pre-neoplastic lymphocytes for acquisition of additional genetic and epigenetic abnormalities that collectively result in neoplastic transformation.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Carioferinas/genética , Leucemia Linfocítica Crónica de Células B/genética , Mutación , Receptores Citoplasmáticos y Nucleares/genética , Animales , Epigénesis Genética , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Modelos Moleculares , Estudios Retrospectivos , Transcriptoma , Proteína Exportina 1
17.
Biochem Soc Trans ; 48(6): 2753-2767, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33300986

RESUMEN

The transport of histones from the cytoplasm to the nucleus of the cell, through the nuclear membrane, is a cellular process that regulates the supply of new histones in the nucleus and is key for DNA replication and transcription. Nuclear import of histones is mediated by proteins of the karyopherin family of nuclear transport receptors. Karyopherins recognize their cargos through linear motifs known as nuclear localization/export sequences or through folded domains in the cargos. Karyopherins interact with nucleoporins, proteins that form the nuclear pore complex, to promote the translocation of their cargos into the nucleus. When binding to histones, karyopherins not only function as nuclear import receptors but also as chaperones, protecting histones from non-specific interactions in the cytoplasm, in the nuclear pore and possibly in the nucleus. Studies have also suggested that karyopherins might participate in histones deposition into nucleosomes. In this review we describe structural and biochemical studies from the last two decades on how karyopherins recognize and transport the core histone proteins H3, H4, H2A and H2B and the linker histone H1 from the cytoplasm to the nucleus, which karyopherin is the major nuclear import receptor for each of these histones, the oligomeric state of histones during nuclear import and the roles of post-translational modifications, histone-chaperones and RanGTP in regulating these nuclear import pathways.


Asunto(s)
Núcleo Celular/metabolismo , Citoplasma/metabolismo , Histonas/metabolismo , Carioferinas/química , Transporte Activo de Núcleo Celular , Proteínas de Ciclo Celular/metabolismo , GTP Fosfohidrolasas/química , Histonas/química , Humanos , Chaperonas Moleculares/metabolismo , Proteínas Nucleares/metabolismo , Conformación Proteica , Procesamiento Proteico-Postraduccional , Receptores Citoplasmáticos y Nucleares/metabolismo , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/metabolismo
18.
Mol Biol Cell ; 31(17): 1879-1891, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32520643

RESUMEN

The E571K mutation of CRM1 is highly prevalent in some cancers, but its mechanism of tumorigenesis is unclear. Glu571 of CRM1 is located in its nuclear export signal (NES)-binding groove, suggesting that binding of select NESs may be altered. We generated HEK 293 cells with either monoallelic CRM1WT/E571K or biallelic CRM1E571K/E571K using CRISPR/Cas9. We also combined analysis of binding affinities and structures of 27 diverse NESs for wild-type and E571K CRM1 with structure-based bioinformatics. While most NESs bind the two CRM1 similarly, NESs from Mek1, eIF4E-transporter, and RPS2 showed >10-fold affinity differences. These NESs have multiple charged side chains binding close to CRM1 position 571, but this feature alone was not sufficient to predict different binding to CRM1(E571K). Consistent with eIF4E-transporter NES binding weaker to CRM1(E571K), eIF4E-transporter was mislocalized in tumor cells carrying CRM1(E571K). This serves as proof of concept that understanding how CRM1(E571K) affects NES binding provides a platform for identifying cargoes that are mislocalized in cancer upon CRM1 mutation. Finally, we showed that large affinity changes seen with some NES peptides (of Mek1 and RPS2) do not always translate to the full-length cargoes, suggesting limitations with current NES prediction methods. Therefore, comprehensive studies like ours are imperative to identify CRM1 cargoes with real pathogenic potential.


Asunto(s)
Carioferinas/genética , Señales de Exportación Nuclear/genética , Receptores Citoplasmáticos y Nucleares/genética , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos/genética , Núcleo Celular/metabolismo , Cristalografía por Rayos X/métodos , Células HEK293 , Humanos , Carioferinas/metabolismo , MAP Quinasa Quinasa 1/metabolismo , Modelos Moleculares , Mutación/genética , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Unión Proteica/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Ribosómicas/metabolismo , Proteína de Unión al GTP ran/genética , Proteína de Unión al GTP ran/metabolismo , Proteína Exportina 1
19.
Curr Opin Cell Biol ; 64: 112-123, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32474299

RESUMEN

Several aggregation-prone RNA-binding proteins, including FUS, EWS, TAF15, hnRNP A1, hnRNP A2, and TDP-43, are mutated in neurodegenerative diseases. The nuclear-cytoplasmic distribution of these proteins is controlled by proteins in the karyopherin family of nuclear transport factors (Kaps). Recent studies have shown that Kaps not only transport these proteins but also inhibit their self-association/aggregation, acting as molecular chaperones. This chaperone activity is impaired for disease-causing mutants of the RNA-binding proteins. Here, we review physical data on the mechanisms of self-association of several disease-associated RNA-binding proteins, through liquid-liquid phase separation and amyloid fiber formation. In each case, we relate these data to biophysical, biochemical, and cell biological data on the inhibition of self-association by Kaps. Our analyses suggest that Kaps may be effective chaperones because they contain large surfaces with diverse physical properties that enable them to engage multiple different regions of their cargo proteins, blocking self-association.


Asunto(s)
beta Carioferinas/metabolismo , Secuencia de Aminoácidos , Núcleo Celular/metabolismo , Metilación de ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Chaperonas Moleculares/metabolismo , beta Carioferinas/química
20.
Bio Protoc ; 10(10): e3625, 2020 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-33659298

RESUMEN

Histones constitute the protein components of nucleosomes. Despite their small sizes, histones do not diffuse through the nuclear pore complex. Instead, they are transported to the nucleus by importins, either alone or in complex with histone chaperones. Determining the molecular size of the importin-histone complexes is key to understanding the mechanism of histone transport and also the potential roles of importins as histone chaperones and in the assembly of nucleosomes. Here we report a simple and reproducible sedimentation-velocity based method to determine the molecular sizes of importin-histone complexes using analytical ultracentrifugation. The method does not use any reporter tags or interaction with column resin thereby analyzing the interactions of the native proteins.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA