Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Genetics ; 217(1): 1-17, 2021 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-33683371

RESUMO

We describe here phase-separated subnuclear organelles in the nematode Caenorhabditis elegans, which we term NUN (NUclear Nervous system-specific) bodies. Unlike other previously described subnuclear organelles, NUN bodies are highly cell type specific. In fully mature animals, 4-10 NUN bodies are observed exclusively in the nucleus of neuronal, glial and neuron-like cells, but not in other somatic cell types. Based on co-localization and genetic loss of function studies, NUN bodies are not related to other previously described subnuclear organelles, such as nucleoli, splicing speckles, paraspeckles, Polycomb bodies, promyelocytic leukemia bodies, gems, stress-induced nuclear bodies, or clastosomes. NUN bodies form immediately after cell cycle exit, before other signs of overt neuronal differentiation and are unaffected by the genetic elimination of transcription factors that control many other aspects of neuronal identity. In one unusual neuron class, the canal-associated neurons, NUN bodies remodel during larval development, and this remodeling depends on the Prd-type homeobox gene ceh-10. In conclusion, we have characterized here a novel subnuclear organelle whose cell type specificity poses the intriguing question of what biochemical process in the nucleus makes all nervous system-associated cells different from cells outside the nervous system.


Assuntos
Espaço Intranuclear/ultraestrutura , Neurônios/ultraestrutura , Animais , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Ciclo Celular , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Espaço Intranuclear/metabolismo , Neuroglia/ultraestrutura
2.
Int J Mol Sci ; 20(12)2019 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-31248120

RESUMO

Polyphosphoinositides (PPIns) are a family of seven lipid messengers that regulate a vast array of signalling pathways to control cell proliferation, migration, survival and differentiation. PPIns are differentially present in various sub-cellular compartments and, through the recruitment and regulation of specific proteins, are key regulators of compartment identity and function. Phosphoinositides and the enzymes that synthesise and degrade them are also present in the nuclear membrane and in nuclear membraneless compartments such as nuclear speckles. Here we discuss how PPIns in the nucleus are modulated in response to external cues and how they function to control downstream signalling. Finally we suggest a role for nuclear PPIns in liquid phase separations that are involved in the formation of membraneless compartments within the nucleus.


Assuntos
Núcleo Celular/metabolismo , Metabolismo dos Lipídeos , Fosfatidilinositóis/metabolismo , Animais , Fenômenos Químicos , Biologia Computacional , Humanos , Espaço Intranuclear/metabolismo , Redes e Vias Metabólicas , Membrana Nuclear/metabolismo , Fosfatidilinositol 4,5-Difosfato/química , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatidilinositóis/química , Transdução de Sinais
3.
Biochem Pharmacol ; 158: 141-152, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30096289

RESUMO

Patients undergoing surgery can suffer from various complications, including post-operative bleeding, local or systematic infection, and neurologic disorders. Major surgery can initiate innate immune responses and trigger overproduction of inflammatory mediators, which can contribute to organ dysfunction. Inflammasomes are innate immune complexes, which are connected to the pathogenesis of various diseases, including atherosclerosis, hemorrhagic brain injury, and Alzheimer's disease. In the present study, we hypothesized that nucleotide-binding oligomerization domain-containing-like receptor protein (NLRP) inflammasomes may have a role in the pathological effects of surgery. Therefore, we designed a protein inhibitor of nuclear factor kappa B (NF-κB) p65 transcripts, called nt-p65-TMD (nuclear transducible (nt) transcription modulated domain (TMD) of RelA (p65)), that can penetrate the nucleus, and evaluated its therapeutic efficacy for dampening surgery-induced inflammasome activation. It was found that the nt-p65-TMD significantly reduced the NLRP1 inflammasome complex components (NLRP1, ASC, and Caspase-1) and interleukin (IL)-1ß and IL-18 productions in the spleen after surgery. In the spleen, specific cell population and selective mediators were altered after surgery with/without nt-p65-TMD treatment. Also, we found that treatment of nt-p65-TMD decreased cell death in the spleen after surgery. Therefore, nt-p65-TMD is a potential novel strategy for reducing surgery-induced NLRP1 inflammasome and complications.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Inflamassomos/metabolismo , Espaço Intranuclear/metabolismo , Complicações Pós-Operatórias/tratamento farmacológico , Complicações Pós-Operatórias/metabolismo , Fator de Transcrição RelA/administração & dosagem , Abdome/cirurgia , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Proteínas Reguladoras de Apoptose/metabolismo , Inflamassomos/antagonistas & inibidores , Intestinos/cirurgia , Espaço Intranuclear/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Complicações Pós-Operatórias/etiologia
4.
Mutat Res ; 809: 99-107, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-28521962

RESUMO

In the nucleus, there are several membraneless structures called nuclear bodies. Among them, promyelocytic leukemia nuclear bodies (PML-NBs) are involved in multiple genome maintenance pathways including the DNA damage response, DNA repair, telomere homeostasis, and p53-associated apoptosis. In response to DNA damage, PML-NBs are coalesced and divided by a fission mechanism, thus increasing their number. PML-NBs also play a role in repairing DNA double-strand breaks (DSBs) by homologous recombination (HR). Clinically, the dominant negative PML-RARα fusion protein expressed in acute promyelocytic leukemia (APL) inhibits the transactivation of downstream factors and disrupts PML function, revealing the tumor suppressor role of PML-NBs. All-trans retinoic acid and arsenic trioxide treatment has been implemented for promyelocytic leukemia to target the PML-RARα fusion protein. PML-NBs are associated with various factors implicated in genome maintenance, and are found at the sites of DNA damage. Their interaction with proteins such as p53 indicates that PML-NBs may play a significant role in apoptosis and cancer. Decades of research have revealed the importance of PML-NBs in diverse cellular pathways, yet the underlying molecular mechanisms and exact functions of PML-NBs remain elusive. In this review, PML protein modifications and the functional relevance of PML-NB and its associated factors in genome maintenance will be discussed.


Assuntos
Reparo do DNA , Instabilidade Genômica , Espaço Intranuclear , Leucemia Promielocítica Aguda , Proteínas de Fusão Oncogênica/metabolismo , Proteína da Leucemia Promielocítica/metabolismo , Animais , Apoptose , Quebras de DNA de Cadeia Dupla , Humanos , Espaço Intranuclear/metabolismo , Espaço Intranuclear/patologia , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patologia , Proteínas de Fusão Oncogênica/genética , Proteína da Leucemia Promielocítica/genética , Homeostase do Telômero
5.
Cancer Sci ; 106(7): 848-56, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25891951

RESUMO

Nucleus accumbens associated 1 (NACC1) is a cancer-associated BTB/POZ (pox virus and zinc finger/bric-a-brac tramtrack broad complex) gene, and is involved in several cellular functions in neurons, cancer and stem cells. Some of the BTB/POZ proteins associated with cancer biology are SUMOylated, which appears to play an important role in transcription regulation. We show that NACC1 is SUMOylated on a phylogenetically conserved lysine (K167) out of three consensus SUMOylation motif sites. Amino acid substitution in the SIM sequence (SIM/M) within the BTB/POZ domain partially reduced K167 SUMOylation activity of NACC1. Overexpression of GFP-NACC1 fusion protein leads to formation of discrete nuclear foci similar to promyelocytic leukemia nuclear bodies (PML-NB), which colocalized with SUMO paralogues (SUMO1/2/3). Both NACC1 nuclear body formation and colocalization with SUMO paralogues were completely suppressed in the GFP-NACC1-SIM/M mutant, whereas they were partially maintained in the NACC1 K167R mutant. Confocal immunofluorescence analysis showed that endogenous and exogenous NACC1 proteins colocalized with endogenous PML protein. A pull-down assay revealed that the consensus motifs of the SUMO acceptor site at K167 and the SIM within the BTB/POZ domain were both necessary for efficient binding to PML protein. Our study demonstrates that NACC1 can be modified by SUMO paralogues, and cooperates with PML protein.


Assuntos
Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Sumoilação , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Sequência de Aminoácidos , Células HeLa , Humanos , Espaço Intranuclear/metabolismo , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patologia , Células MCF-7 , Dados de Sequência Molecular , Proteína da Leucemia Promielocítica , Transporte Proteico
6.
Proc Natl Acad Sci U S A ; 111(8): E776-83, 2014 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-24516149

RESUMO

The mechanisms that control invariant natural killer T (iNKT)-cell development and function are still poorly understood. The mechanistic or mammalian target of rapamycin (mTOR) integrates various environmental signals/cues to regulate cell growth, proliferation, metabolism, and survival. We report here that ablation of mTOR complex 1 (mTORC1) signaling by conditionally deleting Raptor causes severe defects in iNKT-cell development at early stages, leading to drastic reductions in iNKT-cell numbers in the thymus and periphery. In addition, loss of Raptor impairs iNKT-cell proliferation and production of cytokines upon α-galactosylceramide stimulation in vitro and in vivo, and inhibits liver inflammation in an iNKT cell-mediated hepatitis model. Furthermore, Raptor deficiency and rapamycin treatment lead to aberrant intracellular localization and functional impairment of promyelocytic leukemia zinc-finger, a transcription factor critical for iNKT-cell development and effector programs. Our findings define an essential role of mTORC1 to direct iNKT-cell lineage development and effector function.


Assuntos
Diferenciação Celular/imunologia , Fatores de Transcrição Kruppel-Like/imunologia , Complexos Multiproteicos/imunologia , Células T Matadoras Naturais/imunologia , Serina-Treonina Quinases TOR/imunologia , Timócitos/imunologia , Análise de Variância , Animais , Western Blotting , Transplante de Medula Óssea , Bromodesoxiuridina , Morte Celular/imunologia , Proliferação de Células , Imunoprecipitação da Cromatina , Primers do DNA/genética , Citometria de Fluxo , Genes Codificadores dos Receptores de Linfócitos T/genética , Espaço Intranuclear/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Proteína com Dedos de Zinco da Leucemia Promielocítica , Reação em Cadeia da Polimerase em Tempo Real , Estatísticas não Paramétricas , Timócitos/citologia
7.
PLoS One ; 8(6): e65285, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23776465

RESUMO

Nucleolus is a dynamic structure that controls biogenesis of ribosomal RNA and senses cellular stresses. Nucleolus contains a number of proteins including ribosomal proteins that conduct cellular stresses to downstream signaling such as p53 pathway. Recently, it has been reported that modification by a ubiquitin-like molecule, Nedd8, regulates subnuclear localization of ribosomal protein L11. Most of L11 is normally localized and neddylated in nucleolus. However, cellular stress triggers deneddylation and redistribution of L11, and subsequent activation of p53. Although Nedd8 modification is thought to be important for L11 localization, the mechanism of how neddylation of L11 is regulated remains largely unknown. Here, we show that Myeloma overexpressed 2 (Myeov2) controls L11 localization through down-regulation of Nedd8 modification. Expression of Myeov2 reduced neddylation of proteins including L11. We also found that Myeov2 associates with L11 and withholds L11 in nucleoplasm. Although Myeov2 interacted with a Nedd8 deconjugation enzyme COP9 signalosome, L11 deneddylation was mediated by another deneddylase Nedp1, independently of Myeov2. Finally, p53 transcriptional activity is upregulated by Myeov2 expression. These data demonstrate that Myeov2 hampers L11 neddylation through their interactions and confines L11 to nucleoplasm to modulate nucleolar integrity. Our findings provide a novel link between oncogenic stress and p53 pathway and may shed light on the protective mechanism against cancer.


Assuntos
Nucléolo Celular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Espaço Intranuclear/metabolismo , Modelos Biológicos , Proteínas Ribossômicas/metabolismo , Estresse Fisiológico/fisiologia , Ubiquitinas/metabolismo , Primers do DNA/genética , Células HEK293 , Humanos , Immunoblotting , Imuno-Histoquímica , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intercelular , Luciferases , Proteína NEDD8 , Plasmídeos/genética
8.
Retrovirology ; 9: 77, 2012 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-23009398

RESUMO

BACKGROUND: The Tax protein encoded by Human T-lymphotropic virus type 1 (HTLV-1) is a powerful activator of the NF-κB pathway, a property critical for HTLV-1-induced immortalization of CD4⁺ T lymphocytes. Tax permanently stimulates this pathway at a cytoplasmic level by activating the IκB kinase (IKK) complex and at a nuclear level by enhancing the binding of the NF-κB factor RelA to its cognate promoters and by forming nuclear bodies, believed to represent transcriptionally active structures. In previous studies, we reported that Tax ubiquitination and SUMOylation play a critical role in Tax localization and NF-κB activation. Indeed, analysis of lysine Tax mutants fused or not to ubiquitin or SUMO led us to propose a two-step model in which Tax ubiquitination first intervenes to activate IKK while Tax SUMOylation is subsequently required for promoter activation within Tax nuclear bodies. However, recent studies showing that ubiquitin or SUMO can modulate Tax activities in either the nucleus or the cytoplasm and that SUMOylated Tax can serve as substrate for ubiquitination suggested that Tax ubiquitination and SUMOylation may mediate redundant rather than successive functions. RESULTS: In this study, we analyzed the properties of a new Tax mutant that is properly ubiquitinated, but defective for both nuclear body formation and SUMOylation. We report that reducing Tax SUMOylation and nuclear body formation do not alter the ability of Tax to activate IKK, induce RelA nuclear translocation, and trigger gene expression from a NF-κB promoter. Importantly, potent NF-κB promoter activation by Tax despite low SUMOylation and nuclear body formation is also observed in T cells, including CD4⁺ primary T lymphocytes. Moreover, we show that Tax nuclear bodies are hardly observed in HTLV-1-infected T cells. Finally, we provide direct evidence that the degree of NF-κB activation by Tax correlates with the level of Tax ubiquitination, but not SUMOylation. CONCLUSIONS: These data reveal that the formation of Tax nuclear bodies, previously associated to transcriptional activities in Tax-transfected cells, is dispensable for NF-κB promoter activation, notably in CD4⁺ T cells. They also provide the first evidence that Tax SUMOylation is not a key determinant for Tax-induced NF-κB activation.


Assuntos
Produtos do Gene tax/metabolismo , Espaço Intranuclear/metabolismo , NF-kappa B/metabolismo , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional , Ativação Transcricional , Substituição de Aminoácidos , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/virologia , Linhagem Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Produtos do Gene tax/genética , Genes Reporter , Interações Hospedeiro-Patógeno , Vírus Linfotrópico T Tipo 1 Humano/metabolismo , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Humanos , Quinase I-kappa B/metabolismo , Luciferases de Renilla/biossíntese , Luciferases de Renilla/genética , Microscopia Confocal , NF-kappa B/fisiologia , Ligação Proteica , Transporte Proteico , Proteínas Recombinantes de Fusão/metabolismo , Proteína SUMO-1/metabolismo , Transdução de Sinais , Sumoilação , Transcrição Gênica , Ubiquitinação
9.
Proc Natl Acad Sci U S A ; 109(13): 4846-50, 2012 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-22416126

RESUMO

Proteins of the Drosophila behavior/human splicing (DBHS) family include mammalian SFPQ (PSF), NONO (p54nrb), PSPC1, and invertebrate NONA and Hrp65. DBHS proteins are predominately nuclear, and are involved in transcriptional and posttranscriptional gene regulatory functions as well as DNA repair. DBHS proteins influence a wide gamut of biological processes, including the regulation of circadian rhythm, carcinogenesis, and progression of cancer. Additionally, mammalian DBHS proteins associate with the architectural long noncoding RNA NEAT1 (Menε/ß) to form paraspeckles, subnuclear bodies that alter gene expression via the nuclear retention of RNA. Here we describe the crystal structure of the heterodimer of the multidomain conserved region of the DBHS proteins, PSPC1 and NONO. These proteins form an extensively intertwined dimer, consistent with the observation that the different DBHS proteins are typically copurified from mammalian cells, and suggesting that they act as obligate heterodimers. The PSPC1/NONO heterodimer has a right-handed antiparallel coiled-coil that positions two of four RNA recognition motif domains in an unprecedented arrangement on either side of a 20-Å channel. This configuration is supported by a protein:protein interaction involving the NONA/paraspeckle domain, which is characteristic of the DBHS family. By examining various mutants and truncations in cell culture, we find that DBHS proteins require an additional antiparallel coiled-coil emanating from either end of the dimer for paraspeckle subnuclear body formation. These results suggest that paraspeckles may potentially form through self-association of DBHS dimers into higher-order structures.


Assuntos
Espaço Intranuclear/metabolismo , Proteínas Associadas à Matriz Nuclear/química , Proteínas Associadas à Matriz Nuclear/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Fatores de Transcrição de Octâmero/química , Fatores de Transcrição de Octâmero/metabolismo , Multimerização Proteica , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Sequência de Aminoácidos , Sequência Conservada/genética , Proteínas de Ligação a DNA , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Domínios e Motivos de Interação entre Proteínas , Estrutura Terciária de Proteína , Relação Estrutura-Atividade
10.
Traffic ; 13(2): 257-72, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22023725

RESUMO

The reorganization of nuclear structures is an important early feature of apoptosis and involves the activity of specific proteases and nucleases. Well-known is the condensation and fragmentation of chromatin; however, much less is understood about the mechanisms involved in the reorganization of structures from the interchromatin space, such as interchromatin granule clusters (IGCs). In this study, we show that the initial enlargement and rounding-up of IGCs correlate with a decrease in mRNA transcription and are caspase-independent, but involve protein phosphatases PP1/PP2A. Subsequently, multiple enlarged IGCs dissociate from chromatin and fuse into a single structure. The dissociation requires caspase activity and involves caspase-activated DNase (CAD). Apoptotic IMR-5 cells, lacking a proper processing of CAD, show multiple enlarged IGCs that remain linked with chromatin. Overexpression of CAD in IMR-5 cells results in the dissociation of IGCs from chromatin, but the fusion into a single structure remains disturbed. Nuclear matrix protein NuMA is reorganized in a caspase-dependent way around fused IGCs. In conclusion, we show here that the apoptotic rearrangement of IGCs, the nuclear matrix and chromatin are closely associated, occur in defined stages and depend on the activity of protein phosphatases, caspases and CAD.


Assuntos
Antígenos Nucleares/metabolismo , Apoptose/fisiologia , Caspases/metabolismo , Desoxirribonucleases/metabolismo , Proteínas Associadas à Matriz Nuclear/metabolismo , Proteína Fosfatase 2/metabolismo , Ribonucleoproteínas/metabolismo , Spliceossomos/metabolismo , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 7/metabolismo , Caspase 8/metabolismo , Caspase 9/metabolismo , Inibidores de Caspase , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Desoxirribonucleases/genética , Humanos , Espaço Intranuclear/efeitos dos fármacos , Espaço Intranuclear/metabolismo , Espaço Intranuclear/ultraestrutura , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , Proteínas Nucleares/metabolismo , Nucleofosmina , Fosforilação/efeitos dos fármacos , Proteínas de Ligação a Poli-ADP-Ribose , Proteína Fosfatase 1/antagonistas & inibidores , Proteína Fosfatase 1/metabolismo , Proteína Fosfatase 2/antagonistas & inibidores , Ribonucleoproteína Nuclear Pequena U1/metabolismo , Fatores de Processamento de Serina-Arginina , Estaurosporina/farmacologia , Transfecção , Proteínas Centrais de snRNP/metabolismo
11.
J Cell Sci ; 124(Pt 21): 3603-18, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22045732

RESUMO

Telomerase-negative tumor cells use an alternative lengthening of telomeres (ALT) pathway that involves DNA recombination and repair to maintain their proliferative potential. The cytological hallmark of this process is the accumulation of promyelocytic leukemia (PML) nuclear protein at telomeric DNA to form ALT-associated PML bodies (APBs). Here, the de novo formation of a telomeric PML nuclear subcompartment was investigated by recruiting APB protein components. We show that functionally distinct proteins were able to initiate the formation of bona fide APBs with high efficiency in a self-organizing and self-propagating manner. These included: (1) PML and Sp100 as the constituting components of PML nuclear bodies, (2) telomere repeat binding factors 1 and 2 (TRF1 and TRF2, respectively), (3) the DNA repair protein NBS1 and (4) the SUMO E3 ligase MMS21, as well as the isolated SUMO1 domain, through an interacting domain of another protein factor. By contrast, the repair factors Rad9, Rad17 and Rad51 were less efficient in APB nucleation but were recruited to preassembled APBs. The artificially created APBs induced telomeric extension through a DNA repair mechanism, as inferred from their colocalization with sites of non-replicative DNA synthesis and histone H2A.X phosphorylation, and an increase of the telomere repeat length. These activities were absent after recruitment of the APB factors to a pericentric locus and establish APBs as functional intermediates of the ALT pathway.


Assuntos
Corpos de Inclusão Intranuclear/metabolismo , Espaço Intranuclear/metabolismo , Leucemia Promielocítica Aguda/metabolismo , Proteínas Nucleares/metabolismo , Telômero/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Linhagem Celular Tumoral , Humanos , Leucemia Promielocítica Aguda/genética , Proteínas Nucleares/genética , Proteína da Leucemia Promielocítica , Proteína 1 de Ligação a Repetições Teloméricas/genética , Proteína 1 de Ligação a Repetições Teloméricas/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/genética , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética
12.
J Cell Biol ; 194(5): 689-703, 2011 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-21893597

RESUMO

The correlation between stress-induced nucleolar disruption and abrogation of p53 degradation is evident after a wide variety of cellular stresses. This link may be caused by steps in p53 regulation occurring in nucleoli, as suggested by some biochemical evidence. Alternatively, nucleolar disruption also causes redistribution of nucleolar proteins, potentially altering their interactions with p53 and/or MDM2. This raises the fundamental question of whether the nucleolus controls p53 directly, i.e., as a site where p53 regulatory processes occur, or indirectly, i.e., by determining the cellular localization of p53/MDM2-interacting factors. In this work, transport experiments based on heterokaryons, photobleaching, and micronucleation demonstrate that p53 regulatory events are directly regulated by nucleoli and are dependent on intact nucleolar structure and function. Subcellular fractionation and nucleolar isolation revealed a distribution of ubiquitylated p53 that supports these findings. In addition, our results indicate that p53 is exported by two pathways: one stress sensitive and one stress insensitive, the latter being regulated by activities present in the nucleolus.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Nucléolo Celular/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Células 3T3 , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Fusão Celular , Linhagem Celular Tumoral , Nucléolo Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Núcleo Celular/efeitos da radiação , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Cicloeximida/farmacologia , Citoplasma/metabolismo , Dano ao DNA/fisiologia , Dano ao DNA/efeitos da radiação , Demecolcina/farmacologia , Ácidos Graxos Insaturados/farmacologia , Proteínas de Fluorescência Verde/genética , Humanos , Espaço Intranuclear/metabolismo , Leupeptinas/farmacologia , Camundongos , Modelos Biológicos , Região Organizadora do Nucléolo/efeitos dos fármacos , Fotodegradação , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Frações Subcelulares/metabolismo , Proteína Supressora de Tumor p53/genética , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitinação/efeitos dos fármacos , Ubiquitinação/fisiologia
13.
Int J Pharm ; 413(1-2): 254-9, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21536119

RESUMO

Previously, we had reported improving transfection efficiency of the chitosan-plasmid DNA (CS/pDNA)complex via enhancing intracellular unpacking of the exogene by the utilization of phosphorylatable short peptide conjugated chitosan (pSP-CS). In this article, we addressed a novel strategy of nucleus localization signal linked nucleic kinase substrate short peptide (NNS) modification for further optimization of the transfection efficiency. NNS, consisting of "PKKRKVREEAIKFSEEQRFRR", contained a SV40 nucleus localization signal and a potentially phosphorylatable serine residue. The short peptide could be selectively phosphorylated in the nucleus in various mammalian cells. This phosphorylatable NNS (pNNS) was conjugated to chitosan and combined with Cy3 fluorescence labeled plasmid DNA to form a pNNS-CS/pDNA complex. In vitro phosphorylation and DNA releasing assays verified that pNNS could be effectively and selectively phosphorylated by nucleic lysate, hence promoting pDNA unpacking from the complex. Thereafter, C2C12 myoblast cells were transfected. Nuclear localization of the pDNA was represented by the fluorescence in the nucleus and transfection efficiency was determined by the expression of the luciferase reporter gene, which is carried by the plasmid DNA. The results revealed that, compared with lipofactamine2000 and the previously reported pSP-CS, pNNS-CS could transport more pDNA into the nucleus and intensively augment luciferase reporter gene expression. In conclusion, nucleus localization and unpacking from the delivery vector are both critical factors in influencing exogene expression, and pNNS modification is valuable in improving transfection efficacy of the chitosan.


Assuntos
Quitosana/metabolismo , Composição de Medicamentos/métodos , Espaço Intranuclear/metabolismo , Sinais de Localização Nuclear/metabolismo , Transfecção/métodos , Quitosana/química , DNA/metabolismo , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Genes Reporter , Vetores Genéticos , Indicadores e Reagentes/metabolismo , Lipídeos/genética , Lipídeos/farmacologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Peptídeos/análise , Peptídeos/genética , Peptídeos/metabolismo , Fosforilação , Plasmídeos/genética , Plasmídeos/metabolismo
14.
J Cell Biol ; 192(5): 855-71, 2011 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-21383080

RESUMO

Centrosomes are closely associated with the nuclear envelope (NE) throughout the cell cycle and this association is maintained in prophase when they separate to establish the future mitotic spindle. At this stage, the kinetochore constituents CENP-F, NudE, NudEL, dynein, and dynactin accumulate at the NE. We demonstrate here that the N-terminal domain of the nuclear pore complex (NPC) protein Nup133, although largely dispensable for NPC assembly, is required for efficient anchoring of the dynein/dynactin complex to the NE in prophase. Nup133 exerts this function through an interaction network via CENP-F and NudE/EL. We show that this molecular chain is critical for maintaining centrosome association with the NE at mitotic entry and contributes to this process without interfering with the previously described RanBP2-BICD2-dependent pathway of centrosome anchoring. Finally, our study reveals that tethering of centrosomes to the nuclear surface at the G2/M transition contributes, along with other cellular mechanisms, to early stages of bipolar spindle assembly.


Assuntos
Centrossomo/metabolismo , Membrana Nuclear/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/fisiologia , Poro Nuclear/metabolismo , Prófase , Proteínas de Transporte/metabolismo , Proteínas de Transporte/fisiologia , Linhagem Celular Tumoral , Polaridade Celular , Centrossomo/ultraestrutura , Proteínas Cromossômicas não Histona/metabolismo , Proteínas Cromossômicas não Histona/fisiologia , Complexo Dinactina , Dineínas/metabolismo , Células HeLa , Humanos , Espaço Intranuclear/metabolismo , Espaço Intranuclear/ultraestrutura , Proteínas dos Microfilamentos/metabolismo , Proteínas dos Microfilamentos/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Antígenos de Histocompatibilidade Menor , Membrana Nuclear/ultraestrutura , Complexo de Proteínas Formadoras de Poros Nucleares/química , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Mapeamento de Interação de Proteínas , Fuso Acromático/metabolismo
15.
J Cell Biol ; 192(5): 735-50, 2011 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-21357745

RESUMO

Rad54, a member of the SWI/SNF protein family of DNA-dependent ATPases, repairs DNA double-strand breaks (DSBs) through homologous recombination. Here we demonstrate that Rad54 is required for the timely accumulation of the homologous recombination proteins Rad51 and Brca2 at DSBs. Because replication protein A and Nbs1 accumulation is not affected by Rad54 depletion, Rad54 is downstream of DSB resection. Rad54-mediated Rad51 accumulation does not require Rad54's ATPase activity. Thus, our experiments demonstrate that SWI/SNF proteins may have functions independent of their ATPase activity. However, quantitative real-time analysis of Rad54 focus formation indicates that Rad54's ATPase activity is required for the disassociation of Rad54 from DNA and Rad54 turnover at DSBs. Although the non-DNA-bound fraction of Rad54 reversibly interacts with a focus, independent of its ATPase status, the DNA-bound fraction is immobilized in the absence of ATP hydrolysis by Rad54. Finally, we show that ATP hydrolysis by Rad54 is required for the redistribution of DSB repair sites within the nucleus.


Assuntos
Trifosfato de Adenosina/fisiologia , DNA Helicases/fisiologia , Reparo do DNA , Genoma , Proteínas Nucleares/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Células Cultivadas , Quebras de DNA de Cadeia Dupla , DNA Helicases/análise , DNA Helicases/genética , Proteínas de Fluorescência Verde/análise , Espaço Intranuclear/metabolismo , Espaço Intranuclear/ultraestrutura , Camundongos , Proteínas Nucleares/análise , Proteínas Nucleares/genética , Rad51 Recombinase/análise , Rad51 Recombinase/metabolismo , Rad51 Recombinase/fisiologia , Recombinação Genética
16.
Hum Mol Genet ; 20(10): 1873-85, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21330300

RESUMO

The AFF (AF4/FMR2) family of genes includes four members: AFF1/AF4, AFF2/FMR2, AFF3/LAF4 and AFF4/AF5q31. AFF2/FMR2 is silenced in FRAXE intellectual disability, while the other three members have been reported to form fusion genes as a consequence of chromosome translocations with the myeloid/lymphoid or mixed lineage leukemia (MLL) gene in acute lymphoblastic leukemias (ALLs). All AFF proteins are localized in the nucleus and their role as transcriptional activators with a positive action on RNA elongation was primarily studied. We have recently shown that AFF2/FMR2 localizes to nuclear speckles, subnuclear structures considered as storage/modification sites of pre-mRNA splicing factors, and modulates alternative splicing via the interaction with the G-quadruplex RNA-forming structure. We show here that similarly to AFF2/FMR2, AFF3/LAF4 and AFF4/AF5q31 localize to nuclear speckles and are able to bind RNA, having a high apparent affinity for the G-quadruplex structure. Interestingly, AFF3/LAF4 and AFF4/AF5q31, like AFF2/FMR2, modulate, in vivo, the splicing efficiency of a mini-gene containing a G-quadruplex structure in one alternatively spliced exon. Furthermore, we observed that the overexpression of AFF2/3/4 interferes with the organization and/or biogenesis of nuclear speckles. These findings fit well with our observation that enlarged nuclear speckles are present in FRAXE fibroblasts. Furthermore, our findings suggest functional redundancy among the AFF family members in the regulation of splicing and transcription. It is possible that other members of the AFF family compensate for the loss of AFF2/FMR2 activity and as such explain the relatively mild to borderline phenotype observed in FRAXE patients.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Síndrome do Cromossomo X Frágil/genética , Síndrome do Cromossomo X Frágil/patologia , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Fibroblastos/metabolismo , Expressão Gênica/genética , Ordem dos Genes , Genes Reporter/genética , Células HeLa , Humanos , Espaço Intranuclear/metabolismo , Dados de Sequência Molecular , Transporte Proteico , Splicing de RNA/genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
18.
Eukaryot Cell ; 9(3): 379-86, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20081062

RESUMO

Spliceosomal small nuclear ribonucleoproteins (snRNPs) in trypanosomes contain either the canonical heptameric Sm ring or variant Sm cores with snRNA-specific Sm subunits. Here we show biochemically by a combination of RNase H cleavage and tandem affinity purification that the U4 snRNP contains a variant Sm heteroheptamer core in which only SmD3 is replaced by SSm4. This U4-specific, nuclear-localized Sm core protein is essential for growth and splicing. As shown by RNA interference (RNAi) knockdown, SSm4 is specifically required for the integrity of the U4 snRNA and the U4/U6 di-snRNP in trypanosomes. In addition, we demonstrate by in vitro reconstitution of Sm cores that under stringent conditions, the SSm4 protein suffices to specify the assembly of U4 Sm cores. Together, these data indicate that the assembly of the U4-specific Sm core provides an essential step in U4/U6 di-snRNP biogenesis and splicing in trypanosomes.


Assuntos
Multimerização Proteica/fisiologia , Splicing de RNA , RNA Nuclear Pequeno/metabolismo , Ribonucleoproteína Nuclear Pequena U4-U6/biossíntese , Trypanosoma brucei brucei/metabolismo , Proteínas Centrais de snRNP/metabolismo , Proliferação de Células , Centrifugação com Gradiente de Concentração , Expressão Gênica/genética , Espaço Intranuclear/metabolismo , Espectrometria de Massas , Ligação Proteica/genética , Interferência de RNA , RNA de Cadeia Dupla/genética , RNA Nuclear Pequeno/genética , Ribonuclease H/metabolismo , Ribonucleoproteína Nuclear Pequena U4-U6/química , Ribonucleoproteína Nuclear Pequena U4-U6/genética , Trypanosoma brucei brucei/genética , Proteínas Centrais de snRNP/genética
19.
J Biol Chem ; 285(8): 5941-53, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-20018885

RESUMO

p62, also known as sequestosome1 (SQSTM1), A170, or ZIP, is a multifunctional protein implicated in several signal transduction pathways. p62 is induced by various forms of cellular stress, is degraded by autophagy, and acts as a cargo receptor for autophagic degradation of ubiquitinated targets. It is also suggested to shuttle ubiquitinated proteins for proteasomal degradation. p62 is commonly found in cytosolic protein inclusions in patients with protein aggregopathies, it is up-regulated in several forms of human tumors, and mutations in the gene are linked to classical adult onset Paget disease of the bone. To this end, p62 has generally been considered to be a cytosolic protein, and little attention has been paid to possible nuclear roles of this protein. Here, we present evidence that p62 shuttles continuously between nuclear and cytosolic compartments at a high rate. The protein is also found in nuclear promyelocytic leukemia bodies. We show that p62 contains two nuclear localization signals and a nuclear export signal. Our data suggest that the nucleocytoplasmic shuttling of p62 is modulated by phosphorylations at or near the most important nuclear localization signal, NLS2. The aggregation of p62 in cytosolic bodies also regulates the transport of p62 between the compartments. We found p62 to be essential for accumulation of polyubiquitinated proteins in promyelocytic leukemia bodies upon inhibition of nuclear protein export. Furthermore, p62 contributed to the assembly of proteasome-containing degradative compartments in the vicinity of nuclear aggregates containing polyglutamine-expanded Ataxin1Q84 and to the degradation of Ataxin1Q84.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Corpos de Inclusão Intranuclear/metabolismo , Espaço Intranuclear/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Sinais de Localização Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Ubiquitinação , Transporte Ativo do Núcleo Celular/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Ataxina-1 , Ataxinas , Células HeLa , Humanos , Corpos de Inclusão Intranuclear/genética , Proteínas do Tecido Nervoso/genética , Sinais de Localização Nuclear/genética , Proteínas Nucleares/genética , Peptídeos/genética , Peptídeos/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteína Sequestossoma-1
20.
Endocrinology ; 150(11): 4855-62, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19819955

RESUMO

Chronic hyperglycemia causes oxidative stress, which contributes to damage in various tissues and cells, including pancreatic beta-cells. The expression levels of antioxidant enzymes in the islet are low compared with other tissues, rendering the beta-cell more susceptible to damage caused by hyperglycemia. The aim of this study was to investigate whether increasing levels of endogenous glutathione peroxidase-1 (GPx-1), specifically in beta-cells, can protect them against the adverse effects of chronic hyperglycemia and assess mechanisms that may be involved. C57BLKS/J mice overexpressing the antioxidant enzyme GPx-1 only in pancreatic beta-cells were generated. The biological effectiveness of the overexpressed GPx-1 transgene was documented when beta-cells of transgenic mice were protected from streptozotocin. The transgene was then introgressed into the beta-cells of db/db mice. Without use of hypoglycemic agents, hyperglycemia in db/db-GPx(+) mice was initially ameliorated compared with db/db-GPx(-) animals and then substantially reversed by 20 wk of age. beta-Cell volume and insulin granulation and immunostaining were greater in db/db-GPx(+) animals compared with db/db-GPx(-) animals. Importantly, the loss of intranuclear musculoaponeurotic fibrosarcoma oncogene homolog A (MafA) that was observed in nontransgenic db/db mice was prevented by GPx-1 overexpression, making this a likely mechanism for the improved glycemic control. These studies demonstrate that enhancement of intrinsic antioxidant defenses of the beta-cell protects it against deterioration during hyperglycemia.


Assuntos
Diabetes Mellitus/genética , Expressão Gênica , Glutationa Peroxidase/genética , Células Secretoras de Insulina/enzimologia , Espaço Intranuclear/metabolismo , Fatores de Transcrição Maf Maior/metabolismo , Animais , Glicemia , Diabetes Mellitus/enzimologia , Diabetes Mellitus/metabolismo , Modelos Animais de Doenças , Feminino , Glutationa Peroxidase/metabolismo , Humanos , Hiperglicemia/enzimologia , Hiperglicemia/genética , Hiperglicemia/metabolismo , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Glutationa Peroxidase GPX1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA