Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61.344
Filtrar
1.
Cell Stem Cell ; 31(5): 583-585, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38701751

RESUMEN

How nuclear RNA homeostasis impacts cellular functions remains elusive. In this issue of Cell Stem Cell, Han et al.1 utilized a controllable protein degradation system targeting EXOSC2 to perturb RNA homeostasis in mouse pluripotent embryonic stem cells, revealing its vital role in orchestrating crucial nuclear events for cellular fitness.


Asunto(s)
Homeostasis , ARN Nuclear , Animales , Ratones , ARN Nuclear/metabolismo , ARN Nuclear/genética , Complejo Multienzimático de Ribonucleasas del Exosoma/metabolismo , Complejo Multienzimático de Ribonucleasas del Exosoma/genética , Núcleo Celular/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Embrionarias de Ratones/citología , Humanos , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , ARN/metabolismo , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/citología
2.
Nat Commun ; 15(1): 3880, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38719804

RESUMEN

Correlative evidence has suggested that the methyl-CpG-binding protein MeCP2 contributes to the formation of heterochromatin condensates via liquid-liquid phase separation. This interpretation has been reinforced by the observation that heterochromatin, DNA methylation and MeCP2 co-localise within prominent foci in mouse cells. The findings presented here revise this view. MeCP2 localisation is independent of heterochromatin as MeCP2 foci persist even when heterochromatin organisation is disrupted. Additionally, MeCP2 foci fail to show hallmarks of phase separation in live cells. Importantly, we find that mouse cellular models are highly atypical as MeCP2 distribution is diffuse in most mammalian species, including humans. Notably, MeCP2 foci are absent in Mus spretus which is a mouse subspecies lacking methylated satellite DNA repeats. We conclude that MeCP2 has no intrinsic tendency to form condensates and its localisation is independent of heterochromatin. Instead, the distribution of MeCP2 in the nucleus is primarily determined by global DNA methylation patterns.


Asunto(s)
Metilación de ADN , Heterocromatina , Proteína 2 de Unión a Metil-CpG , Proteína 2 de Unión a Metil-CpG/metabolismo , Proteína 2 de Unión a Metil-CpG/genética , Heterocromatina/metabolismo , Animales , Ratones , Humanos , Núcleo Celular/metabolismo , Unión Proteica , ADN/metabolismo , ADN Satélite/metabolismo , ADN Satélite/genética , Separación de Fases
3.
J Cell Biol ; 223(6)2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38709216

RESUMEN

Autophagy is an essential degradation program required for cell homeostasis. Among its functions is the engulfment and destruction of cytosolic pathogens, termed xenophagy. Not surprisingly, many pathogens use various strategies to circumvent or co-opt autophagic degradation. For poxviruses, it is known that infection activates autophagy, which however is not required for successful replication. Even though these complex viruses replicate exclusively in the cytoplasm, autophagy-mediated control of poxvirus infection has not been extensively explored. Using the prototypic poxvirus, vaccinia virus (VACV), we show that overexpression of the xenophagy receptors p62, NDP52, and Tax1Bp1 restricts poxvirus infection. While NDP52 and Tax1Bp1 were degraded, p62 initially targeted cytoplasmic virions before being shunted to the nucleus. Nuclear translocation of p62 was dependent upon p62 NLS2 and correlated with VACV kinase mediated phosphorylation of p62 T269/S272. This suggests that VACV targets p62 during the early stages of infection to avoid destruction and further implies that poxviruses exhibit multi-layered control of autophagy to facilitate cytoplasmic replication.


Asunto(s)
Autofagia , Núcleo Celular , Proteína Sequestosoma-1 , Virus Vaccinia , Humanos , Transporte Activo de Núcleo Celular , Núcleo Celular/metabolismo , Núcleo Celular/virología , Células HEK293 , Células HeLa , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Fosforilación , Proteína Sequestosoma-1/metabolismo , Proteína Sequestosoma-1/genética , Vaccinia/metabolismo , Vaccinia/virología , Vaccinia/genética , Virus Vaccinia/metabolismo , Virus Vaccinia/genética , Replicación Viral
4.
Nucleus ; 15(1): 2349085, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38700207

RESUMEN

The ESCRT machinery plays a pivotal role in membrane-remodeling events across multiple cellular processes including nuclear envelope repair and reformation, nuclear pore complex surveillance, endolysosomal trafficking, and neuronal pruning. Alterations in ESCRT-III functionality have been associated with neurodegenerative diseases including Frontotemporal Dementia (FTD), Amyotrophic Lateral Sclerosis (ALS), and Alzheimer's Disease (AD). In addition, mutations in specific ESCRT-III proteins have been identified in FTD/ALS. Thus, understanding how disruptions in the fundamental functions of this pathway and its individual protein components in the human central nervous system (CNS) may offer valuable insights into mechanisms underlying neurodegenerative disease pathogenesis and identification of potential therapeutic targets. In this review, we discuss ESCRT components, dynamics, and functions, with a focus on the ESCRT-III pathway. In addition, we explore the implications of altered ESCRT-III function for neurodegeneration with a primary emphasis on nuclear surveillance and endolysosomal trafficking within the CNS.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte , Enfermedades Neurodegenerativas , Humanos , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/genética , Animales , Núcleo Celular/metabolismo , Demencia Frontotemporal/metabolismo , Demencia Frontotemporal/patología , Demencia Frontotemporal/genética , Endosomas/metabolismo
5.
PLoS One ; 19(5): e0295971, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38709794

RESUMEN

The human genome is pervasively transcribed and produces a wide variety of long non-coding RNAs (lncRNAs), constituting the majority of transcripts across human cell types. Some specific nuclear lncRNAs have been shown to be important regulatory components acting locally. As RNA-chromatin interaction and Hi-C chromatin conformation data showed that chromatin interactions of nuclear lncRNAs are determined by the local chromatin 3D conformation, we used Hi-C data to identify potential target genes of lncRNAs. RNA-protein interaction data suggested that nuclear lncRNAs act as scaffolds to recruit regulatory proteins to target promoters and enhancers. Nuclear lncRNAs may therefore play a role in directing regulatory factors to locations spatially close to the lncRNA gene. We provide the analysis results through an interactive visualization web portal at https://fantom.gsc.riken.jp/zenbu/reports/#F6_3D_lncRNA.


Asunto(s)
Cromatina , ARN Largo no Codificante , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Cromatina/metabolismo , Cromatina/genética , Humanos , Anotación de Secuencia Molecular , Núcleo Celular/metabolismo , Núcleo Celular/genética , Genoma Humano , Regiones Promotoras Genéticas
6.
Sci Adv ; 10(18): eadl6082, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38701207

RESUMEN

The AAA+-ATPase valosin-containing protein (VCP; also called p97 or Cdc48), a major protein unfolding machinery with a variety of essential functions, localizes to different subcellular compartments where it has different functions. However, the processes regulating the distribution of VCP between the cytosol and nucleus are not understood. Here, we identified p37 (also called UBXN2B) as a major factor regulating VCP nucleocytoplasmic shuttling. p37-dependent VCP localization was crucial for local cytosolic VCP functions, such as autophagy, and nuclear functions in DNA damage repair. Mutations in VCP causing multisystem proteinopathy enhanced its association with p37, leading to decreased nuclear localization of VCP, which enhanced susceptibility to DNA damage accumulation. Both VCP localization and DNA damage susceptibility in cells with such mutations were normalized by lowering p37 levels. Thus, we uncovered a mechanism by which VCP nucleocytoplasmic distribution is fine-tuned, providing a means for VCP to respond appropriately to local needs.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Núcleo Celular , Citosol , Proteína que Contiene Valosina , Proteína que Contiene Valosina/metabolismo , Proteína que Contiene Valosina/genética , Humanos , Citosol/metabolismo , Núcleo Celular/metabolismo , Mutación , Transporte Activo de Núcleo Celular , Daño del ADN , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfatasas/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Transporte de Proteínas , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Reparación del ADN , Autofagia , Unión Proteica , Células HEK293
7.
Int J Biol Sci ; 20(7): 2748-2762, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38725859

RESUMEN

Abnormal nuclear enlargement is a diagnostic and physical hallmark of malignant tumors. Large nuclei are positively associated with an increased risk of developing metastasis; however, a large nucleus is inevitably more resistant to cell migration due to its size. The present study demonstrated that the nuclear size of primary colorectal cancer (CRC) cells at an advanced stage was larger than cells at an early stage. In addition, the nuclei of CRC liver metastases were larger than those of the corresponding primary CRC tissues. CRC cells were sorted into large-nucleated cells (LNCs) and small-nucleated cells (SNCs). Purified LNCs exhibited greater constricted migratory and metastatic capacity than SNCs in vitro and in vivo. Mechanistically, ErbB4 was highly expressed in LNCs, which phosphorylated lamin A/C at serine 22 via the ErbB4-Akt1 signaling pathway. Furthermore, the level of phosphorylated lamin A/C was a negative determinant of nuclear stiffness. Taken together, CRC LNCs possessed greater constricted migratory and metastatic potential than SNCs due to ErbB4-Akt1-mediated lamin A/C phosphorylation and nuclear softening. These results may provide a potential treatment strategy for tumor metastasis by targeting nuclear stiffness in patients with cancer, particularly CRC.


Asunto(s)
Neoplasias Colorrectales , Lamina Tipo A , Proteínas Proto-Oncogénicas c-akt , Receptor ErbB-4 , Transducción de Señal , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Humanos , Receptor ErbB-4/metabolismo , Receptor ErbB-4/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Lamina Tipo A/metabolismo , Animales , Línea Celular Tumoral , Ratones , Núcleo Celular/metabolismo , Movimiento Celular , Masculino , Femenino , Fosforilación , Metástasis de la Neoplasia , Ratones Desnudos
8.
Nat Commun ; 15(1): 3901, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38724505

RESUMEN

Activation of the NF-κB pathway is strictly regulated to prevent excessive inflammatory and immune responses. In a well-known negative feedback model, IκBα-dependent NF-κB termination is a delayed response pattern in the later stage of activation, and the mechanisms mediating the rapid termination of active NF-κB remain unclear. Here, we showed IκBα-independent rapid termination of nuclear NF-κB mediated by CLK2, which negatively regulated active NF-κB by phosphorylating the RelA/p65 subunit of NF-κB at Ser180 in the nucleus to limit its transcriptional activation through degradation and nuclear export. Depletion of CLK2 increased the production of inflammatory cytokines, reduced viral replication and increased the survival of the mice. Mechanistically, CLK2 phosphorylated RelA/p65 at Ser180 in the nucleus, leading to ubiquitin‒proteasome-mediated degradation and cytoplasmic redistribution. Importantly, a CLK2 inhibitor promoted cytokine production, reduced viral replication, and accelerated murine psoriasis. This study revealed an IκBα-independent mechanism of early-stage termination of NF-κB in which phosphorylated Ser180 RelA/p65 turned off posttranslational modifications associated with transcriptional activation, ultimately resulting in the degradation and nuclear export of RelA/p65 to inhibit excessive inflammatory activation. Our findings showed that the phosphorylation of RelA/p65 at Ser180 in the nucleus inhibits early-stage NF-κB activation, thereby mediating the negative regulation of NF-κB.


Asunto(s)
Citoplasma , Inhibidor NF-kappaB alfa , FN-kappa B , Proteínas Tirosina Quinasas , Factor de Transcripción ReIA , Animales , Fosforilación , Inhibidor NF-kappaB alfa/metabolismo , Inhibidor NF-kappaB alfa/genética , Ratones , Factor de Transcripción ReIA/metabolismo , Humanos , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas/genética , FN-kappa B/metabolismo , Citoplasma/metabolismo , Proteolisis , Núcleo Celular/metabolismo , Replicación Viral , Células HEK293 , Transducción de Señal , Ratones Endogámicos C57BL , Citocinas/metabolismo , Transporte Activo de Núcleo Celular , Proteínas Serina-Treonina Quinasas
9.
J Nanobiotechnology ; 22(1): 261, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38760744

RESUMEN

Delayed repair of fractures seriously impacts patients' health and significantly increases financial burdens. Consequently, there is a growing clinical demand for effective fracture treatment. While current materials used for fracture repair have partially addressed bone integrity issues, they still possess limitations. These challenges include issues associated with autologous material donor sites, intricate preparation procedures for artificial biomaterials, suboptimal biocompatibility, and extended degradation cycles, all of which are detrimental to bone regeneration. Hence, there is an urgent need to design a novel material with a straightforward preparation method that can substantially enhance bone regeneration. In this context, we developed a novel nanoparticle, mPPTMP195, to enhance the bioavailability of TMP195 for fracture treatment. Our results demonstrate that mPPTMP195 effectively promotes the differentiation of bone marrow mesenchymal stem cells into osteoblasts while inhibiting the differentiation of bone marrow mononuclear macrophages into osteoclasts. Moreover, in a mouse femur fracture model, mPPTMP195 nanoparticles exhibited superior therapeutic effects compared to free TMP195. Ultimately, our study highlights that mPPTMP195 accelerates fracture repair by preventing HDAC4 translocation from the cytoplasm to the nucleus, thereby activating the NRF2/HO-1 signaling pathway. In conclusion, our study not only proposes a new strategy for fracture treatment but also provides an efficient nano-delivery system for the widespread application of TMP195 in various other diseases.


Asunto(s)
Diferenciación Celular , Histona Desacetilasas , Células Madre Mesenquimatosas , Nanopartículas , Animales , Ratones , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Nanopartículas/química , Diferenciación Celular/efectos de los fármacos , Histona Desacetilasas/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Ratones Endogámicos C57BL , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteoblastos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Hemo-Oxigenasa 1/metabolismo , Masculino , Regeneración Ósea/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Núcleo Celular/metabolismo , Curación de Fractura/efectos de los fármacos , Humanos , Proteínas de la Membrana
10.
Nucleus ; 15(1): 2350182, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38738760

RESUMEN

Long noncoding RNAs (LncRNAs) are key regulators of gene expression and can mediate their effects in both the nucleus and cytoplasm. Some of the best-characterized lncRNAs are localized within the nucleus, where they modulate the nuclear architecture and influence gene expression. In this review, we discuss the role of lncRNAs in nuclear architecture in the context of their gene regulatory functions in innate immunity. Here, we discuss various approaches to functionally characterize nuclear-localized lncRNAs and the challenges faced in the field.


Asunto(s)
Núcleo Celular , ARN Largo no Codificante , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Humanos , Núcleo Celular/metabolismo , Núcleo Celular/genética , Animales , Inmunidad Innata , Regulación de la Expresión Génica
11.
PLoS Comput Biol ; 20(5): e1011416, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38739641

RESUMEN

During meiosis, pairing of homologous chromosomes (homologs) ensures the formation of haploid gametes from diploid precursor cells, a prerequisite for sexual reproduction. Pairing during meiotic prophase I facilitates crossover recombination and homolog segregation during the ensuing reductional cell division. Mechanisms that ensure stable homolog alignment in the presence of an excess of non-homologous chromosomes have remained elusive, but rapid chromosome movements appear to play a role in the process. Apart from homolog attraction, provided by early intermediates of homologous recombination, dissociation of non-homologous associations also appears to contribute to homolog pairing, as suggested by the detection of stable non-homologous chromosome associations in pairing-defective mutants. Here, we have developed an agent-based model for homolog pairing derived from the dynamics of a naturally occurring chromosome ensemble. The model simulates unidirectional chromosome movements, as well as collision dynamics determined by attractive and repulsive forces arising from close-range physical interactions. Chromosome number and size as well as movement velocity and repulsive forces are identified as key factors in the kinetics and efficiency of homologous pairing in addition to homolog attraction. Dissociation of interactions between non-homologous chromosomes may contribute to pairing by crowding homologs into a limited nuclear area thus creating preconditions for close-range homolog attraction. Incorporating natural chromosome lengths, the model accurately recapitulates efficiency and kinetics of homolog pairing observed for wild-type and mutant meiosis in budding yeast, and can be adapted to nuclear dimensions and chromosome sets of other organisms.


Asunto(s)
Emparejamiento Cromosómico , Meiosis , Meiosis/genética , Emparejamiento Cromosómico/genética , Modelos Genéticos , Saccharomyces cerevisiae/genética , Cromosomas Fúngicos/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Simulación por Computador , Biología Computacional
12.
Hum Mol Genet ; 33(R1): R34-R41, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38779776

RESUMEN

In human cells, the nuclear and mitochondrial genomes engage in a complex interplay to produce dual-encoded oxidative phosphorylation (OXPHOS) complexes. The coordination of these dynamic gene expression processes is essential for producing matched amounts of OXPHOS protein subunits. This review focuses on our current understanding of the mitochondrial central dogma rates, highlighting the striking differences in gene expression rates between mitochondrial and nuclear genes. We synthesize a coherent model of mitochondrial gene expression kinetics, highlighting the emerging principles and emphasizing where more precise measurements would be beneficial. Such an understanding is pivotal for grasping the unique aspects of mitochondrial function and its role in cellular energetics, and it has profound implications for aging, metabolic disorders, and neurodegenerative diseases.


Asunto(s)
Mitocondrias , Fosforilación Oxidativa , Humanos , Mitocondrias/genética , Mitocondrias/metabolismo , Regulación de la Expresión Génica , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Genoma Mitocondrial , Metabolismo Energético/genética , Núcleo Celular/metabolismo , Núcleo Celular/genética , Envejecimiento/genética , Envejecimiento/metabolismo , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo
13.
Proc Natl Acad Sci U S A ; 121(22): e2314166121, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38768348

RESUMEN

The nonstructural protein 1 (Nsp1) of SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) is a virulence factor that targets multiple cellular pathways to inhibit host gene expression and antiviral response. However, the underlying mechanisms of the various Nsp1-mediated functions and their contributions to SARS-CoV-2 virulence remain unclear. Among the targets of Nsp1 is the mRNA (messenger ribonucleic acid) export receptor NXF1-NXT1, which mediates nuclear export of mRNAs from the nucleus to the cytoplasm. Based on Nsp1 crystal structure, we generated mutants on Nsp1 surfaces and identified an acidic N-terminal patch that is critical for interaction with NXF1-NXT1. Photoactivatable Nsp1 probe reveals the RNA Recognition Motif (RRM) domain of NXF1 as an Nsp1 N-terminal binding site. By mutating the Nsp1 N-terminal acidic patch, we identified a separation-of-function mutant of Nsp1 that retains its translation inhibitory function but substantially loses its interaction with NXF1 and reverts Nsp1-mediated mRNA export inhibition. We then generated a recombinant (r)SARS-CoV-2 mutant on the Nsp1 N-terminal acidic patch and found that this surface is key to promote NXF1 binding and inhibition of host mRNA nuclear export, viral replication, and pathogenicity in vivo. Thus, these findings provide a mechanistic understanding of Nsp1-mediated mRNA export inhibition and establish the importance of this pathway in the virulence of SARS-CoV-2.


Asunto(s)
Transporte Activo de Núcleo Celular , COVID-19 , Proteínas de Transporte Nucleocitoplasmático , ARN Mensajero , Proteínas de Unión al ARN , SARS-CoV-2 , Proteínas no Estructurales Virales , Humanos , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidad , SARS-CoV-2/genética , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/química , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Proteínas de Transporte Nucleocitoplasmático/genética , Animales , COVID-19/virología , COVID-19/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Replicación Viral , Núcleo Celular/metabolismo , Células Vero , Virulencia , Chlorocebus aethiops , Células HEK293
14.
Genome Biol ; 25(1): 121, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38741206

RESUMEN

Multiomic droplet-based technologies allow different molecular modalities, such as chromatin accessibility and gene expression (scATAC-seq and scRNA-seq), to be probed in the same nucleus. We develop EmptyDropsMultiome, an approach that distinguishes true nuclei-containing droplets from background. Using simulations, we show that EmptyDropsMultiome has higher statistical power and accuracy than existing approaches, including CellRanger-arc and EmptyDrops. On real datasets, we observe that CellRanger-arc misses more than half of the nuclei identified by EmptyDropsMultiome and, moreover, is biased against certain cell types, some of which have a retrieval rate lower than 20%.


Asunto(s)
Análisis de la Célula Individual , Análisis de la Célula Individual/métodos , Humanos , Núcleo Celular/genética , Núcleo Celular/metabolismo , Cromatina/metabolismo , Cromatina/genética , Multiómica
15.
Nucleus ; 15(1): 2352203, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38780365

RESUMEN

In eukaryotic cells, the nuclear envelope (NE) is a membrane partition between the nucleus and the cytoplasm to compartmentalize nuclear contents. It plays an important role in facilitating nuclear functions including transcription, DNA replication and repair. In mammalian cells, the NE breaks down and then reforms during cell division, and in interphase it is restored shortly after the NE rupture induced by mechanical force. In this way, the partitioning effect is regulated through dynamic processes throughout the cell cycle. A failure in rebuilding the NE structure triggers the mixing of nuclear and cytoplasmic contents, leading to catastrophic consequences for the nuclear functions. Whereas the precise details of molecular mechanisms for NE reformation during cell division and NE restoration in interphase are still being investigated, here, we mostly focus on mammalian cells to describe key aspects that have been identified and to discuss the crosstalk between them.


Asunto(s)
Mitosis , Membrana Nuclear , Membrana Nuclear/metabolismo , Humanos , Animales , Reparación del ADN , Núcleo Celular/metabolismo
16.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167224, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38723872

RESUMEN

BACKGROUND: Pentamethylquercetin (PMQ) is a natural polymethyl flavonoid that possesses anti-apoptotic and other biological properties. Abdominal aortic aneurysm (AAA), a fatal vascular disease with a high risk of rupture, is associated with phenotypic switching and apoptosis of medial vascular smooth muscle cells (VSMCs). This study aimed to investigate the protective effects of PMQ on the development of AAA and the underlying mechanism. METHODS: ApoE-/- mice were continuously infused with angiotensin II (Ang II) for 4 weeks to develop the AAA model. Intragastric administration of PMQ was initiated 5 days before Ang II infusion and continued for 4 weeks. In vitro, VSMCs were cultured and pretreated with PMQ, stimulated with Ang II. Real-time PCR, western blotting, and immunofluorescence staining were used to examine the roles and mechanisms of PMQ on the phenotypic switching and apoptosis of VSMCs. RESULTS: PMQ dose-dependently reduced the incidence of Ang II-induced AAA, aneurysm diameter enlargement, elastin degradation, VSMCs phenotypic switching and apoptosis. Furthermore, PMQ also inhibited phenotypic switching and apoptosis in Ang II-stimulated VSMCs. PMQ exerted protective effects by regulating the C/EBPß/PTEN/AKT/GSK-3ß axis. AAV-mediated overexpression of PTEN reduced the therapeutic effects of PMQ in the AAA model mice, suggesting that the effects of PMQ on Ang II-mediated AAA formation were related to the PTEN/AKT/GSK-3ß axis. PMQ inhibited VSMCs phenotypic switching and apoptosis by bounding to C/EBPß at Lys253 with hydrogen bond to regulate C/EBPß nuclear translocation and PTEN/AKT/GSK-3ß axis, thereby inhibiting Ang II-induced AAA formation. CONCLUSIONS: Pentamethylquercetin inhibits angiotensin II-induced abdominal aortic aneurysm formation by bounding to C/EBPß at Lys253. Therefore, PMQ prevents the formation of AAA and reduces the incidence of AAA.


Asunto(s)
Angiotensina II , Aneurisma de la Aorta Abdominal , Apoptosis , Músculo Liso Vascular , Quercetina , Animales , Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/prevención & control , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/tratamiento farmacológico , Angiotensina II/farmacología , Ratones , Quercetina/análogos & derivados , Quercetina/farmacología , Apoptosis/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Masculino , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Modelos Animales de Enfermedad , Fosfohidrolasa PTEN/metabolismo , Fosfohidrolasa PTEN/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratones Endogámicos C57BL , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Transducción de Señal/efectos de los fármacos , Células Cultivadas , Núcleo Celular/metabolismo , Núcleo Celular/efectos de los fármacos
17.
Curr Protoc ; 4(5): e1042, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38767195

RESUMEN

Biochemical fractionation is a technique used to isolate and separate distinct cellular compartments, critical for dissecting cellular mechanisms and molecular pathways. Herein we outline a biochemical fraction methodology for isolation of ultra-pure nuclei and cytoplasm. This protocol utilizes hypotonic lysis buffer to suspend cells, coupled with a calibrated centrifugation strategy, for enhanced separation of cytoplasm from the nuclear fraction. Subsequent purification steps ensure the integrity of the isolated nuclear fraction. Overall, this method facilitates accurate protein localization, essential for functional studies, demonstrating its efficacy in separating cellular compartments. © 2024 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol: Biochemical fractionation Support Protocol 1: Protein quantification using Bradford assay Support Protocol 2: SDS/PAGE and Western blotting.


Asunto(s)
Fraccionamiento Celular , Núcleo Celular , Citoplasma , Citoplasma/metabolismo , Citoplasma/química , Núcleo Celular/metabolismo , Núcleo Celular/química , Fraccionamiento Celular/métodos , Humanos , Electroforesis en Gel de Poliacrilamida , Western Blotting
18.
Cells ; 13(9)2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38727317

RESUMEN

mTOR is a central regulator of cell growth and metabolism in response to mitogenic and nutrient signals. Notably, mTOR is not only found in the cytoplasm but also in the nucleus. This review highlights direct involvement of nuclear mTOR in regulating transcription factors, orchestrating epigenetic modifications, and facilitating chromatin remodeling. These effects intricately modulate gene expression programs associated with growth and metabolic processes. Furthermore, the review underscores the importance of nuclear mTOR in mediating the interplay between metabolism and epigenetic modifications. By integrating its functions in nutrient signaling and gene expression related to growth and metabolism, nuclear mTOR emerges as a central hub governing cellular homeostasis, malignant transformation, and cancer progression. Better understanding of nuclear mTOR signaling has the potential to lead to novel therapies against cancer and other growth-related diseases.


Asunto(s)
Núcleo Celular , Proliferación Celular , Transducción de Señal , Serina-Treonina Quinasas TOR , Humanos , Serina-Treonina Quinasas TOR/metabolismo , Núcleo Celular/metabolismo , Animales , Epigénesis Genética , Transcripción Genética , Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/patología
19.
Mol Cell ; 84(9): 1635-1636, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38701740

RESUMEN

In a recent publication in Nature, Xu et al.1 discovered a role of CRL5-SPSB3 ubiquitin ligase in promoting ubiquitination and degradation of nuclear cGAS, which prevents aberrant cGAS activation by genomic DNA and contributes to the maintenance of immune homeostasis.


Asunto(s)
Homeostasis , Nucleotidiltransferasas , Ubiquitinación , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/genética , Humanos , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Núcleo Celular/metabolismo , Proteolisis , Animales
20.
FASEB J ; 38(10): e23629, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38742770

RESUMEN

The molecular and cellular basis of health in human tendons remains poorly understood. Among human tendons, hamstring tendon has markedly low pathology and can provide a prototypic healthy tendon reference. The aim of this study was to determine the transcriptomes and location of all cell types in healthy hamstring tendon. Using single nucleus RNA sequencing, we profiled the transcriptomes of 10 533 nuclei from four healthy donors and identified 12 distinct cell types. We confirmed the presence of two fibroblast cell types, endothelial cells, mural cells, and immune cells, and identified cell types previously unreported in tendons, including different skeletal muscle cell types, satellite cells, adipocytes, and undefined nervous system cells. The location of these cell types within tendon was defined using spatial transcriptomics and imaging, and potential transcriptional networks and cell-cell interactions were analyzed. We demonstrate that fibroblasts have the highest number of potential cell-cell interactions in our dataset, are present throughout the tendon, and play an important role in the production and organization of extracellular matrix, thus confirming their role as key regulators of hamstring tendon homeostasis. Overall, our findings underscore the complexity of the cellular networks that underpin healthy human tendon function and the central role of fibroblasts as key regulators of hamstring tendon tissue homeostasis.


Asunto(s)
Perfilación de la Expresión Génica , Tendones Isquiotibiales , Transcriptoma , Humanos , Masculino , Adulto , Tendones Isquiotibiales/metabolismo , Fibroblastos/metabolismo , Femenino , Núcleo Celular/metabolismo , Núcleo Celular/genética , Matriz Extracelular/metabolismo , Tendones/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA