Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 12(1): 1911, 2022 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-35115587

RESUMO

Many critical advances in research utilize techniques that combine high-resolution with high-content characterization at the single cell level. We introduce the MICS (MACSima Imaging Cyclic Staining) technology, which enables the immunofluorescent imaging of hundreds of protein targets across a single specimen at subcellular resolution. MICS is based on cycles of staining, imaging, and erasure, using photobleaching of fluorescent labels of recombinant antibodies (REAfinity Antibodies), or release of antibodies (REAlease Antibodies) or their labels (REAdye_lease Antibodies). Multimarker analysis can identify potential targets for immune therapy against solid tumors. With MICS we analysed human glioblastoma, ovarian and pancreatic carcinoma, and 16 healthy tissues, identifying the pair EPCAM/THY1 as a potential target for chimeric antigen receptor (CAR) T cell therapy for ovarian carcinoma. Using an Adapter CAR T cell approach, we show selective killing of cells only if both markers are expressed. MICS represents a new high-content microscopy methodology widely applicable for personalized medicine.


Assuntos
Biomarcadores Tumorais/metabolismo , Molécula de Adesão da Célula Epitelial/metabolismo , Imunofluorescência , Imunoterapia Adotiva , Neoplasias/metabolismo , Neoplasias/terapia , Fotodegradação , Análise de Célula Única , Antígenos Thy-1/metabolismo , Morte Celular , Citotoxicidade Imunológica , Ensaios de Triagem em Larga Escala , Humanos , Neoplasias/imunologia , Neoplasias/patologia , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/transplante
2.
Glia ; 65(6): 990-1004, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28317180

RESUMO

Astrocytes are the most abundant cell type of the central nervous system and cover a broad range of functionalities. We report here the generation of a novel monoclonal antibody, anti-astrocyte cell surface antigen-2 (Anti-ACSA-2). Flow cytometry, immunohistochemistry and immunocytochemistry revealed that Anti-ACSA-2 reacted specifically with a not yet identified glycosylated surface molecule of murine astrocytes at all developmental stages. It did not show any labeling of non-astroglial cells such as neurons, oligodendrocytes, NG2+ cells, microglia, endothelial cells, leukocytes, or erythrocytes. Co-labeling studies of GLAST and ACSA-2 showed largely overlapping expression. However, there were also notable differences in protein expression levels and frequencies of single-positive subpopulations of cells in some regions of the CNS such as cerebellum, most prominently at early postnatal stages. In the neurogenic niches, the dentate gyrus of the hippocampus and the subventricular zone (SVZ), again a general overlap with slight differences in expression levels were observed. ACSA-2 was unlike GLAST not sensitive to papain-based tissue dissociation and allowed for a highly effective, acute, specific, and prospective purification of viable astrocytes based on a new rapid sorting procedure using Anti-ACSA-2 directly coupled to superparamagnetic MicroBeads. In conclusion, ACSA-2 appears to be a new surface marker for astrocytes, radial glia, neural stem cells and bipotent glial progenitor cells which opens up the possibility of further dissecting the characteristics of astroglial subpopulations and lineages.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos de Superfície/análise , Antígenos de Superfície/imunologia , Astrócitos/citologia , Astrócitos/imunologia , Separação Imunomagnética/métodos , Animais , Animais Recém-Nascidos , Especificidade de Anticorpos , Antígenos de Superfície/metabolismo , Encéfalo/citologia , Encéfalo/crescimento & desenvolvimento , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/imunologia , Eritrócitos/citologia , Eritrócitos/metabolismo , Transportador 1 de Aminoácido Excitatório/análise , Leucócitos/citologia , Leucócitos/imunologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/citologia , Microglia/imunologia , Células-Tronco Neurais/imunologia , Neurônios/citologia , Neurônios/metabolismo , Oligodendroglia/citologia , Oligodendroglia/imunologia , Ratos Wistar
3.
Arch Toxicol ; 91(2): 827-837, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27116294

RESUMO

Food supplements based on herbal products are widely used during pregnancy as part of a self-care approach. The idea that such supplements are safe and healthy is deeply seated in the general population, although they do not underlie the same strict safety regulations than medical drugs. We aimed to characterize the neurodevelopmental effects of the green tea catechin epigallocatechin gallate (EGCG), which is now commercialized as high-dose food supplement. We used the "Neurosphere Assay" to study the effects and unravel underlying molecular mechanisms of EGCG treatment on human and rat neural progenitor cells (NPCs) development in vitro. EGCG alters human and rat NPC development in vitro. It disturbs migration distance, migration pattern, and nuclear density of NPCs growing as neurospheres. These functional impairments are initiated by EGCG binding to the extracellular matrix glycoprotein laminin, preventing its binding to ß1-integrin subunits, thereby prohibiting cell adhesion and resulting in altered glia alignment and decreased number of migrating young neurons. Our data raise a concern on the intake of high-dose EGCG food supplements during pregnancy and highlight the need of an in vivo characterization of the effects of high-dose EGCG exposure during neurodevelopment.


Assuntos
Catequina/análogos & derivados , Células-Tronco Neurais/efeitos dos fármacos , Animais , Catequina/administração & dosagem , Catequina/efeitos adversos , Catequina/metabolismo , Catequina/farmacologia , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Suplementos Nutricionais , Feminino , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Integrina beta1/metabolismo , Laminina/metabolismo , Nestina/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Gravidez , Ratos
4.
Nucleic Acids Res ; 43(19): 9123-32, 2015 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-26446989

RESUMO

The in vivo potency of antisense oligonucleotides (ASO) has been significantly increased by reducing their length to 8-15 nucleotides and by the incorporation of high affinity RNA binders such as 2', 4'-bridged nucleic acids (also known as locked nucleic acid or LNA, and 2',4'-constrained ethyl [cET]). We now report the development of a novel ASO design in which such short ASO monomers to one or more targets are co-synthesized as homo- or heterodimers or multimers via phosphodiester linkers that are stable in plasma, but cleaved inside cells, releasing the active ASO monomers. Compared to current ASOs, these multimers and multi-targeting oligonucleotides (MTOs) provide increased plasma protein binding and biodistribution to liver, and increased in vivo efficacy against single or multiple targets with a single construct. In vivo, MTOs synthesized in both RNase H-activating and steric-blocking oligonucleotide designs provide ≈4-5-fold increased potency and ≈2-fold increased efficacy, suggesting broad therapeutic applications.


Assuntos
Oligonucleotídeos Antissenso/química , Animais , Apolipoproteína C-III/genética , Apolipoproteína C-III/metabolismo , Apolipoproteínas B/genética , Apolipoproteínas B/metabolismo , Dimerização , Feminino , Humanos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , MicroRNAs/antagonistas & inibidores , Oligonucleotídeos Antissenso/farmacocinética , Oligonucleotídeos Antissenso/farmacologia , Distribuição Tecidual
5.
Mol Ther ; 19(12): 2163-8, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21971426

RESUMO

Antisense oligonucleotides (ASOs) and small interfering RNA (siRNA) promise specific correction of disease-causing gene expression. Therapeutic implementation, however, has been forestalled by poor delivery to the appropriate tissue, cell type, and subcellular compartment. Topical administration is considered to circumvent these issues. The availability of inhalation devices and unmet medical need in lung disease has focused efforts in this tissue. We report the development of a novel cell sorting method for quantitative, cell type-specific analysis of siRNA, and locked nucleic acid (LNA) ASO uptake and efficacy after intratracheal (i.t.) administration in mice. Through fluorescent dye labeling, we compare the utility of this approach to whole animal and whole tissue analysis, and examine the extent of tissue distribution. We detail rapid systemic access and renal clearance for both therapeutic classes and lack of efficacy at the protein level in lung macrophages, epithelia, or other cell types. We nevertheless observe efficient redirection of i.t. administered phosphorothioate (PS) LNA ASO to the liver and kidney leading to targeted gene knockdown. These data suggest delivery remains a key obstacle to topically administered, naked oligonucleotide efficacy in the lung and introduce inhalation as a potentially viable alternative to injection for antisense administration to the liver and kidneys.


Assuntos
Apolipoproteínas B/antagonistas & inibidores , Inativação Gênica , Luciferases/antagonistas & inibidores , Oligonucleotídeos Antissenso/farmacocinética , Oligonucleotídeos/farmacocinética , RNA Interferente Pequeno/genética , Administração por Inalação , Animais , Apolipoproteínas B/genética , Apolipoproteínas B/metabolismo , Células Cultivadas , Marcação de Genes , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Rim/citologia , Rim/metabolismo , Fígado/citologia , Fígado/metabolismo , Luciferases/genética , Luciferases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Oligonucleotídeos/administração & dosagem , Oligonucleotídeos Antissenso/administração & dosagem , Distribuição Tecidual
6.
Cell Mol Life Sci ; 68(12): 2089-99, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20957508

RESUMO

Neuronal migration is one of the most critical processes during early brain development. The gaseous messenger nitric oxide (NO) has been shown to modulate neuronal and glial migration in various experimental models. Here, we analyze a potential role for NO signaling in the migration of fetal human neural progenitor cells. Cells migrate out of cultured neurospheres and differentiate into both neuronal and glial cells. The neurosphere cultures express neuronal nitric oxide synthase and soluble guanylyl cyclase that produces cGMP upon activation with NO. By employing small bioactive enzyme activators and inhibitors in both gain and loss of function experiments, we show NO/cGMP signaling as a positive regulator of migration in neurosphere cultures of early developing human brain cells. Since NO signaling regulates cell movements from developing insects to mammalian nervous systems, this transduction pathway may have evolutionary conserved functions.


Assuntos
Movimento Celular , GMP Cíclico/metabolismo , Óxido Nítrico/metabolismo , Transdução de Sinais/fisiologia , Células-Tronco/citologia , Técnicas de Cultura de Células , Diferenciação Celular , Humanos , Neuroglia/citologia , Neurônios/citologia
7.
Environ Health Perspect ; 118(11): 1571-7, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20570779

RESUMO

BACKGROUND: Because of their lipophilicity, persistent organic pollutants (POPs) cross the human placenta, possibly affecting central nervous system development. Most POPs are known aryl hydrocarbon receptor (AhR) ligands and activators of AhR signaling. Therefore, AhR activation has been suggested to cause developmental neurotoxicity (DNT). OBJECTIVE: We studied the effects of AhR ligands on basic processes of brain development in two comparative in vitro systems to determine whether AhR-activation is the underlying mechanism for reported DNT of POPs in humans. METHODS: We employed neurosphere cultures based on human neural progenitor cells (hNPCs) and wild-type and AhR-deficient mouse NPCs (mNPCs) and studied the effects of different AhR agonists [3-methylcholanthrene (3-MC), benzo(a)pyrene [B(a)P], and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)] and an antagonist [3'-methoxy-4'-nitroflavone (MNF)] on neurosphere development. Moreover, we analyzed expression of AhR and genes involved in AhR signaling. RESULTS: In contrast to wild-type mNPCs, hNPCs and AhR-deficient mNPCs were insensitive to AhR agonism or antagonism. Although AhR modulation attenuated wild-type mNPC proliferation and migration, hNPCs and AhR-deficient mNPCs remained unaffected. Results also suggest that species-specific differences resulted from nonfunctional AhR signaling in hNPCs. CONCLUSION: Our findings suggest that in contrast to wild-type mNPCs, hNPCs were protected against polycyclic aromatic hydrocarbon-induced DNT because of an absence of AhR This difference may contribute to species-specific differences in sensitivity to POPs.


Assuntos
Poluentes Ambientais/toxicidade , Neurônios/efeitos dos fármacos , Hidrocarbonetos Policíclicos Aromáticos/toxicidade , Receptores de Hidrocarboneto Arílico/metabolismo , Células-Tronco/efeitos dos fármacos , Animais , Benzo(a)pireno/toxicidade , Células Cultivadas , Expressão Gênica/efeitos dos fármacos , Humanos , Metilcolantreno/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Sistema Nervoso/efeitos dos fármacos , Sistema Nervoso/crescimento & desenvolvimento , Sistema Nervoso/metabolismo , Neurônios/metabolismo , Dibenzodioxinas Policloradas/toxicidade , RNA Mensageiro/metabolismo , Receptores de Hidrocarboneto Arílico/genética , Transdução de Sinais/efeitos dos fármacos , Especificidade da Espécie , Células-Tronco/metabolismo
8.
Environ Health Perspect ; 117(7): 1131-8, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19654924

RESUMO

BACKGROUND: Developmental neurotoxicity (DNT) of environmental chemicals is a serious threat to human health. Current DNT testing guidelines propose investigations in rodents, which require large numbers of animals. With regard to the "3 Rs" (reduction, replacement, and refinement) of animal testing and the European regulation of chemicals [Registration, Evaluation, and Authorisation of Chemicals (REACH)], alternative testing strategies are needed in order to refine and reduce animal experiments and allow faster and less expensive screening. OBJECTIVES: The goal of this study was to establish a three-dimensional test system for DNT screening based on human fetal brain cells. METHODS: We established assays suitable for detecting disturbances in basic processes of brain development by employing human neural progenitor cells (hNPCs), which grow as neurospheres. Furthermore, we assessed effects of mercury and oxidative stress on these cells. RESULTS: We found that human neurospheres imitate proliferation, differentiation, and migration in vitro. Exposure to the proapoptotic agent staurosporine further suggests that human neurospheres possess functioning apoptosis machinery. The developmental neurotoxicants methylmercury chloride and mercury chloride decreased migration distance and number of neuronal-like cells in differentiated hNPCs. Furthermore, hNPCs undergo caspase-independent apoptosis when exposed toward high amounts of oxidative stress. CONCLUSIONS: Human neurospheres are likely to imitate basic processes of brain development, and these processes can be modulated by developmental neurotoxicants. Thus, this three-dimensional cell system is a promising approach for DNT testing.


Assuntos
Neurogênese/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Encéfalo/citologia , Encéfalo/embriologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Feto/citologia , Feto/embriologia , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Mercúrio/toxicidade , Compostos de Metilmercúrio/toxicidade
9.
Methods Mol Biol ; 463: 191-202, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18951170

RESUMO

The 20S proteasome is part of a larger complex, the 26S proteasome, that is implicated in the ATP-dependent degradation of multiubiquitin-conjugated proteins (1). About 80% of intracellular protein breakdown occurs via the ubiquitin-proteasome system (UPS). Key proteins such as transcription factors, nuclear receptors, cyclins, cyclin-dependent kinase inhibitors, p53, and NF-kappaB are regulated by this pathway. Thus, the UPS has been implicated to play a role in multiple cellular events including the cell cycle, signal transduction, antigen presentation, and DNA repair and transcription (2, 3). In 1984 Varshavsky and co-workers discovered that ubiquitin-dependent pathways play a role in cell cycle control, and suggested that protein degradation is instrumental in regulation of gene expression (4). Consistent with this idea, Franke and colleagues had shown that proteasomes localize to the nuclei of Xenopus laevis oocytes and HeLa cells (5, 6). Subsequent work confirmed that (i) all components of the UPS that are required for protein degradation indeed reside in the cell nucleus (7); (ii) nuclear proteins are substrates for proteasomal degradation (8); and (iii) proteasome-dependent proteolysis occurs in distinct nucleoplasmic foci (9). The intricate balance between nuclear function and quality control through proteolysis is exemplified by reports that show a correlation of aberrant nuclear protein aggregates with inhibition of transcription in neurodegenerative diseases such as Huntington's chorea and animal and cell culture models of polyglutamine repeat disorders (10,11).Considering the central role of the UPS in nuclear processes, a detailed knowledge of the time and place at which a substrate is ubiquitinylated and degraded will be essential to our understanding of the cellular mechanisms that orchestrate the expression of thousands of genes or development of subnuclear pathologies. Here, we describe fluorescence-based localization methods for proteasomes, protein aggregates, and proteasomal proteolysis in the cell nucleus that may aid to analyse the UPS in housekeeping and disease conditions.


Assuntos
Núcleo Celular/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/química , Células HeLa , Humanos , Imuno-Histoquímica/métodos , Microscopia Confocal/métodos , Microscopia de Fluorescência/métodos , Modelos Biológicos , Nanopartículas/química , Nanotecnologia/métodos , Proteínas Nucleares/metabolismo , Controle de Qualidade
10.
Histochem Cell Biol ; 127(6): 591-601, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17205305

RESUMO

Proteasomes are ATP-driven, multisubunit proteolytic machines that degrade endogenous proteins into peptides and play a crucial role in cellular events such as the cell cycle, signal transduction, maintenance of proper protein folding and gene expression. Recent evidence indicates that the ubiquitin-proteasome system is an active component of the cell nucleus. A characteristic feature of the nucleus is its organization into distinct domains that have a unique composition of macromolecules and dynamically form as a response to the requirements of nuclear function. Here, we show by systematic application of different immunocytochemical procedures and comparison with signature proteins of nuclear domains that during interphase endogenous proteasomes are localized diffusely throughout the nucleoplasm, in speckles, in nuclear bodies, and in nucleoplasmic foci. Proteasomes do not occur in the nuclear envelope region or the nucleolus, unless nucleoplasmic invaginations expand into this nuclear body. Confirmedly, proteasomal proteolysis is detected in nucleoplasmic foci, but is absent from the nuclear envelope or nucleolus. The results underpin the idea that the ubiquitin-proteasome system is not only located, but also proteolytically active in distinct nuclear domains and thus may be directly involved in gene expression, and nuclear quality control.


Assuntos
Biomarcadores/metabolismo , Núcleo Celular/metabolismo , Hepatócitos/metabolismo , Imuno-Histoquímica/métodos , Interfase/fisiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/patologia , Inibidores de Cisteína Proteinase/farmacologia , Técnica Indireta de Fluorescência para Anticorpo , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Humanos , Processamento de Imagem Assistida por Computador , Interfase/efeitos dos fármacos , Microscopia Confocal , Fixação de Tecidos/métodos
11.
J Cell Sci ; 118(Pt 22): 5231-42, 2005 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16249232

RESUMO

The ubiquitin proteasome system plays a fundamental role in the regulation of cellular processes by degradation of endogenous proteins. Proteasomes are localized in both, the cytoplasm and the cell nucleus, however, little is known about nuclear proteolysis. Here, fluorogenic precursor substrates enabled detection of proteasomal activity in nucleoplasmic cell fractions (turnover 0.0541 microM/minute) and nuclei of living cells (turnover 0.0472 microM/minute). By contrast, cell fractions of nucleoli or nuclear envelopes did not contain proteasomal activity. Microinjection of ectopic fluorogenic protein DQ-ovalbumin revealed that proteasomal protein degradation occurs in distinct nucleoplasmic foci, which partially overlap with signature proteins of subnuclear domains, such as splicing speckles or promyelocytic leukemia bodies, ubiquitin, nucleoplasmic proteasomes and RNA polymerase II. Our results establish proteasomal proteolysis as an intrinsic function of the cell nucleus.


Assuntos
Estruturas do Núcleo Celular/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas/metabolismo , Animais , Estruturas do Núcleo Celular/enzimologia , Sobrevivência Celular , Proteínas Cromossômicas não Histona/metabolismo , Células Epiteliais/citologia , Fibroblastos/citologia , Humanos , Queratinócitos/citologia , Lamina Tipo A/metabolismo , Lamina Tipo B/metabolismo , Camundongos , Inibidores de Proteassoma , Tubulina (Proteína)/metabolismo , Ubiquitina/metabolismo
12.
J Struct Biol ; 140(1-3): 189-99, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12490167

RESUMO

Although proteasomes are abundant in the nucleoplasm little is known of proteasome-dependent proteolysis within the nucleus. Thus, we monitored the subcellular distribution of nuclear proteins in correlation with proteasomes. The proteasomal pathway clears away endogenous proteins, regulates numerous cellular processes, and delivers immunocompetent peptides to the antigen presenting machinery. Confocal laser scanning microscopy revealed that histones, splicing factor SC35, spliceosomal components, such as U1-70k or SmB/B('), and PML partially colocalize with 20S proteasomes in nucleoplasmic substructures, whereas the centromeric and nucleolar proteins topoisomerase I, fibrillarin, and UBF did not overlap with proteasomes. The specific inhibition of proteasomal processing with lactacystin induced accumulation of histone protein H2A, SC35, spliceosomal components, and PML, suggesting that these proteins are normally degraded by proteasomes. In contrast, concentrations of centromeric proteins CENP-B and -C and nucleolar proteins remained constant during inhibition of proteasomes. Quantification of fluorescence intensities corroborated that nuclear proteins which colocalize with proteasomes are degraded by proteasome-dependent proteolysis within the nucleoplasm. These data provide evidence that the proteasome proteolytic pathway is involved in processing of nuclear components, and thus may play an important role in the regulation of nuclear structure and function.


Assuntos
Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Núcleo Celular/metabolismo , Cisteína Endopeptidases/metabolismo , Complexos Multienzimáticos/metabolismo , Proteínas Nucleares , Actinas/metabolismo , Sobrevivência Celular , Inibidores de Cisteína Proteinase/farmacologia , Eletroforese em Gel de Poliacrilamida , Células HeLa , Humanos , Immunoblotting , Microscopia Confocal , Microscopia de Fluorescência , Proteínas de Neoplasias/metabolismo , Proteína da Leucemia Promielocítica , Complexo de Endopeptidases do Proteassoma , Spliceossomos/metabolismo , Fatores de Tempo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor
13.
Mol Biol Cell ; 13(10): 3576-87, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12388758

RESUMO

A prerequisite for proteins to interact in a cell is that they are present in the same intracellular compartment. Although it is generally accepted that proteasomes occur in both, the cytoplasm and the nucleus, research has been focusing on cytoplasmic protein breakdown and antigen processing, respectively. Thus, little is known on the functional organization of the proteasome in the nucleus. Here we report that within the nucleus 20S and 26S proteasomes occur throughout the nucleoplasm and partially colocalize with splicing factor-containing speckles. Because proteasomes are absent from the nucleolus, a recruitment system was used to analyze the molecular fate of nucleolar protein fibrillarin: Subtoxic concentrations of mercuric chloride (HgCl(2)) induce subcellular redistribution of fibrillarin and substantial colocalization (33%) with nucleoplasmic proteasomes in different cell lines and in primary cells isolated from mercury-treated mice. Accumulation of fibrillarin and fibrillarin-ubiquitin conjugates in lactacystin-treated cells suggests that proteasome-dependent processing of this autoantigen occurs upon mercury induction. The latter observation might constitute the cell biological basis of autoimmune responses that specifically target fibrillarin in mercury-mouse models and scleroderma.


Assuntos
Acetilcisteína/análogos & derivados , Apresentação de Antígeno , Autoantígenos/metabolismo , Núcleo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Cisteína Endopeptidases/metabolismo , Complexos Multienzimáticos/metabolismo , Peptídeo Hidrolases/metabolismo , Acetilcisteína/metabolismo , Animais , Autoantígenos/genética , Linhagem Celular , Proteínas Cromossômicas não Histona/genética , Inibidores de Cisteína Proteinase/metabolismo , Fosfatases de Especificidade Dupla , Feminino , Humanos , Cloreto de Mercúrio/administração & dosagem , Cloreto de Mercúrio/farmacologia , Camundongos , Fosfoproteínas/metabolismo , Complexo de Endopeptidases do Proteassoma , Proteínas Tirosina Fosfatases/metabolismo , Ratos , Baço/citologia , Baço/metabolismo , Ubiquitina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA