Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Analyst ; 148(8): 1682-1693, 2023 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-36912705

RESUMO

The spatiotemporal visualization of intracellular microRNA (miRNA) plays a critical role in the diagnosis and treatment of malignant disease. Although DNAzyme-based biosensing has been regarded as the most promising candidate, inefficient analytical resolution is frequently encountered. Here, we propose a bioorthogonal approach toward high-fidelity imaging of intracellular miRNA by designing a multifunctional nanoprobe that integrates MnO2 nanosheet-mediated intracellular delivery and activation by a fat mass and obesity-associated protein (FTO)-switched positive feedback. MnO2 nanosheets facilitate nanoprobe delivery and intracellular DNAzyme cofactors are released upon glutathione-triggered reduction. Meanwhile, an m6A-caged DNAzyme probe could be bioorthogonally activated by intracellular FTO to eliminate potential off-target activation. Therefore, the activated DNAzyme probe and substrate probe could recognize miRNA to perform cascade signal amplification in the initiation of the release of Mn2+ from MnO2 nanosheets. This strategy realized high-fidelity imaging of intracellular aberrant miRNA within tumor cells with a satisfactory detection limit of 9.7 pM, paving the way to facilitate clinical tumor diagnosis and prognosis monitoring.


Assuntos
DNA Catalítico , MicroRNAs , Neoplasias , Humanos , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Diagnóstico por Imagem , DNA Catalítico/genética , Compostos de Manganês , MicroRNAs/genética , Neoplasias/patologia , Óxidos , Espaço Intracelular/genética
2.
Immunology ; 163(3): 323-337, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33548066

RESUMO

We have previously identified that human Ku70, a nuclear protein, serves as a cytosolic DNA sensor. Upon transfection with DNA or infection with DNA virus, Ku70 translocates from the nucleus into the cytoplasm and then predominately induces interferon lambda1 (IFN-λ1) rather than IFN-alpha or IFN-beta, through a STING-dependent signalling pathway. However, a detailed mechanism for Ku70 cytoplasmic translocation and its correlation with IFN-λ1 induction have not been fully elucidated. Here, we observed that cytoplasmic translocation of Ku70 only occurred in DNA-triggered IFN-λ1-inducible cells. Additionally, infection by Herpes simplex virus type-1 (HSV-1), a DNA virus, induces cytoplasmic translocation of Ku70 and IFN-λ1 induction in a strain-dependent manner: the translocation and IFN-λ1 induction were detected upon infection by HSV-1 McKrae, but not MacIntyre, strain. A kinetic analysis indicated that cytoplasmic translocation of Ku70 was initiated right after DNA transfection and was peaked at 6 hr after DNA stimulation. Furthermore, treatment with leptomycin B, a nuclear export inhibitor, inhibited both Ku70 translocation and IFN-λ1 induction, suggesting that Ku70 translocation is an essential and early event for its cytosolic DNA sensing. We further confirmed that enhancing the acetylation status of the cells promotes Ku70's cytoplasmic accumulation, and therefore increases DNA-mediated IFN-λ1 induction. These findings provide insights into the molecular mechanism by which the versatile sensor detects pathogenic DNA in a localization-dependent manner.


Assuntos
Citoplasma/metabolismo , Herpes Simples/imunologia , Herpesvirus Humano 1/fisiologia , Interferons/metabolismo , Autoantígeno Ku/metabolismo , Acetilação , Antibióticos Antineoplásicos/farmacologia , DNA Viral/genética , DNA Viral/imunologia , Ácidos Graxos Insaturados/farmacologia , Células HEK293 , Humanos , Interferons/genética , Espaço Intracelular/genética , Espaço Intracelular/imunologia , Transporte Proteico , Especificidade da Espécie , Regulação para Cima
3.
IEEE Trans Nanobioscience ; 19(3): 556-561, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32340955

RESUMO

Due to technological advances the quality and availability of biological data has increased dramatically in the last decade. Analysing protein-protein interaction networks (PPINs) in an integrated way, together with subcellular compartment data, provides such biological context, helps to fill in the gaps between a single type of biological data and genes causing diseases and can identify novel genes related to disease. In this study, we present BCCGD, a method for integrating subcellular localization data with PPINs that detects breast cancer candidate genes in protein complexes. We achieve this by defining the significance of the compartment, constructing edge-weighted PPINs, finding protein complexes with a non-negative matrix factorization approach, generating disease-specific networks based on the known disease genes, prioritizing disease candidate genes with a WDC method. As a case study, we investigate the breast cancer but the techniques described here are applicable to other disorders. For the top genes scored by BCCGD approach, we utilize the literature retrieving method to test the correlations of them with the breast cancer. The results show that BCCGD discover some novel breast cancer candidate genes which are valuable references for the biomedical scientists.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Espaço Intracelular/genética , Mapeamento de Interação de Proteínas/métodos , Mapas de Interação de Proteínas/genética , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/metabolismo , Biologia Computacional , Bases de Dados Factuais , Feminino , Humanos , Espaço Intracelular/metabolismo
4.
Artigo em Inglês | MEDLINE | ID: mdl-30763245

RESUMO

Cancer in essence is a complex genomic alteration disease which is caused by the somatic mutations during the lifetime. According to previous researches, the first step to overcome cancer is to identify driver genes which can promote carcinogenesis. However, it is still a big challenge to precisely and efficiently extract the cancer related driver genes because the nature of cancer is heterogeneous and there exists tremendously irrelevant passenger mutations which have no function impact on the cancer's development. In this work, we proposed a novel entropy-based method namely EntroRank to identify driver genes by integrating the subcellular localization information and mutual exclusive of variation frequency into the network. EntroRank can take into full consideration different properties of driver genes. Considering the modularity of driver genes, the mutated genes in the network were first clustered into different subgroups according to their located compartments. After that, the structural entropy of the gene in the subgroup was employed to measure its indispensability. Considering mutual exclusive property between driver genes in the modules, relative entropy was utilized to measure the degree of mutual exclusive between two mutated genes in terms of their variation frequency. We applied our method to three different cancers including lung, prostate, and breast cancer. The results show our method not only detect the well-known important drivers but also prioritiz the rare unknown driver genes. Besides, EntroRank can identify driver genes having mutual exclusive property. Compared with other existing methods, our method achieves a better performance for most of cancer types in terms of Precision, Recall, and Fscore.


Assuntos
Biologia Computacional/métodos , Genes Neoplásicos/genética , Neoplasias/genética , Carcinogênese/genética , Entropia , Humanos , Espaço Intracelular/genética , Espaço Intracelular/metabolismo , Neoplasias/metabolismo , Transdução de Sinais/genética
5.
Cancer Res ; 79(23): 5901-5906, 2019 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-31431458

RESUMO

In the March 1 issue of Cancer Research, we identified the Wnt receptor Fzd7 as an attractive therapeutic target for the treatment of gastric cancer. In summary, we showed that pharmacological inhibition of Wnt receptors, or genetic deletion of Fzd7, blocks the initiation and growth of gastric tumors. Inhibiting Fzd receptors, specifically Fzd7, inhibits the growth of gastric cancer cells even in the presence of adenomatous polyposis coli (Apc) mutation. Apc is located in the cytoplasm downstream of Fzd7 in the Wnt signaling cascade and APC mutations activate Wnt/ß-catenin signaling, therefore, this result seems counterintuitive. Here, we analyze this result in greater detail in the context of current knowledge of Wnt signaling and discuss the wider implications of this aspect of Wnt signaling in other cancers.


Assuntos
Heterogeneidade Genética , Neoplasias/patologia , Via de Sinalização Wnt/genética , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Membrana Celular/efeitos dos fármacos , Membrana Celular/genética , Membrana Celular/metabolismo , Receptores Frizzled/antagonistas & inibidores , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/genética , Espaço Intracelular/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/genética , Via de Sinalização Wnt/efeitos dos fármacos
6.
Neurochem Res ; 42(1): 92-107, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27422263

RESUMO

Mammalian glutamate dehydrogenase1 (GDH1) (E.C. 1.4.1.3) is a mitochondrial enzyme that catalyzes the reversible oxidative deamination of glutamate to α-ketoglutarate and ammonia while reducing NAD+ and/or NADP+ to NADH and/or NADPH. It links amino acid with carbohydrate metabolism, contributing to Krebs cycle anaplerosis, energy production, ammonia handling and redox homeostasis. Although GDH1 was one of the first major metabolic enzymes to be studied decades ago, its role in cell biology is still incompletely understood. There is however growing interest in a novel GDH2 isoenzyme that emerged via duplication in primates and underwent rapid evolutionary selection concomitant with prefrontal human cortex expansion. Also, the anaplerotic function of GDH1 and GDH2 is currently under sharp focus as this relates to the biology of glial tumors and other neoplasias. Here we used antibodies specific for human GDH1 (hGDH1) and human GDH2 (hGDH2) to study the expression of these isoenzymes in human tissues. Results revealed that both hGDH1 and hGDH2 are expressed in human brain, kidney, testis and steroidogenic organs. However, distinct hGDH1 and hGDH2 expression patterns emerged. Thus, while the Sertoli cells of human testis were strongly positive for hGDH2, they were negative for hGDH1. Conversely, hGDH1 showed very high levels of expression in human liver, but hepatocytes were virtually devoid of hGDH2. In human adrenals, both hGDHs were densely expressed in steroid-producing cells, with hGDH2 expression pattern matching that of the cholesterol side chain cleavage system involved in steroid synthesis. Similarly in human ovaries and placenta, both hGDH1 and hGDH2 were densely expressed in estrogen producing cells. In addition, hGDH1, being a housekeeping enzyme, was also expressed in cells that lack endocrine function. Regarding human brain, study of cortical sections using immunofluorescence (IF) with confocal microscopy revealed that hGDH1 and hGDH2 were both expressed in the cytoplasm of gray and white matter astrocytes within coarse structures resembling mitochondria. Additionally, hGDH1 localized to the nuclear membrane of a subpopulation of astrocytes and of the vast majority of oligodendrocytes and their precursors. Remarkably, hGDH2-specific staining was detected in human cortical neurons, with different expression patterns having emerged. One pattern, observed in large cortical neurons (some with pyramidal morphology), was a hGDH2-specific labeling of cytoplasmic structures resembling mitochondria. These were distributed either in the cell body-axon or on the cell surface in close proximity to astrocytic end-feet that encircle glutamatergic synapses. Another pattern was observed in small cortical neurons with round dense nuclei in which the hGDH2-specific staining was found in the nuclear membrane. A detailed description of these observations and their functional implications, suggesting that the GDH flux is used by different cells to serve some of their unique functions, is presented below.


Assuntos
Corpo Celular/enzimologia , Regulação Enzimológica da Expressão Gênica , Glutamato Desidrogenase/biossíntese , Espaço Intracelular/enzimologia , Sequência de Aminoácidos , Encéfalo/enzimologia , Corpo Celular/genética , Glutamato Desidrogenase/genética , Humanos , Espaço Intracelular/genética , Rim/enzimologia , Fígado/enzimologia , Masculino , Testículo/enzimologia
7.
J Neuropathol Exp Neurol ; 75(8): 779-790, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27346750

RESUMO

Genome-wide association studies have established BIN1 (Bridging Integrator 1) as the most significant late-onset Alzheimer disease (AD) susceptibility locus after APOE We analyzed BIN1 protein expression using automated immunohistochemistry on the hippocampal CA1 region in 19 patients with either no, mild, or moderate-to-marked AD pathology, who had been assessed by Clinical Dementia Rating and CERAD scores. We also examined the amygdala, prefrontal, temporal, and occipital regions in a subset of these patients. In non-demented controls without AD pathology, BIN1 protein was expressed in white matter, glia, particularly oligodendrocytes, and in the neuropil in which the BIN1 signal decorated axons. With increasing severity of AD, BIN1 in the CA1 region showed: 1) sustained expression in glial cells, 2) decreased areas of neuropil expression, and 3) increased cytoplasmic neuronal expression that did not correlate with neurofibrillary tangle load. In patients with AD, both the prefrontal cortex and CA1 showed a decrease in BIN1-immunoreactive (BIN1-ir) neuropil areas and increases in numbers of BIN1-ir neurons. The numbers of CA1 BIN1-ir pyramidal neurons correlated with hippocampal CERAD neuritic plaque scores; BIN1 neuropil signal was absent in neuritic plaques. Our data provide novel insight into the relationship between BIN1 protein expression and the progression of AD-associated pathology and its diagnostic hallmarks.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Doença de Alzheimer/metabolismo , Córtex Cerebral/metabolismo , Progressão da Doença , Proteínas Nucleares/biossíntese , Proteínas Supressoras de Tumor/biossíntese , Proteínas Adaptadoras de Transdução de Sinal/genética , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Córtex Cerebral/patologia , Feminino , Expressão Gênica , Humanos , Espaço Intracelular/genética , Espaço Intracelular/metabolismo , Masculino , Pessoa de Meia-Idade , Proteínas Nucleares/genética , Proteínas Supressoras de Tumor/genética
8.
J Neurosci ; 35(48): 15996-6011, 2015 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-26631479

RESUMO

It is fast emerging that maintaining mitochondrial function is important for regulating astrocyte function, although the specific mechanisms that govern astrocyte mitochondrial trafficking and positioning remain poorly understood. The mitochondrial Rho-GTPase 1 protein (Miro1) regulates mitochondrial trafficking and detachment from the microtubule transport network to control activity-dependent mitochondrial positioning in neurons. However, whether Miro proteins are important for regulating signaling-dependent mitochondrial dynamics in astrocytic processes remains unclear. Using live-cell confocal microscopy of rat organotypic hippocampal slices, we find that enhancing neuronal activity induces transient mitochondrial remodeling in astrocytes, with a concomitant, transient reduction in mitochondrial trafficking, mediated by elevations in intracellular Ca(2+). Stimulating neuronal activity also induced mitochondrial confinement within astrocytic processes in close proximity to synapses. Furthermore, we show that the Ca(2+)-sensing EF-hand domains of Miro1 are important for regulating mitochondrial trafficking in astrocytes and required for activity-driven mitochondrial confinement near synapses. Additionally, activity-dependent mitochondrial positioning by Miro1 reciprocally regulates the levels of intracellular Ca(2+) in astrocytic processes. Thus, the regulation of intracellular Ca(2+) signaling, dependent on Miro1-mediated mitochondrial positioning, could have important consequences for astrocyte Ca(2+) wave propagation, gliotransmission, and ultimately neuronal function.


Assuntos
Astrócitos/ultraestrutura , Sinalização do Cálcio/fisiologia , Espaço Intracelular/metabolismo , Mitocôndrias/fisiologia , Proteínas Mitocondriais/metabolismo , Sinapses/fisiologia , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Dependovirus/genética , Embrião de Mamíferos , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Ácido Glutâmico/farmacologia , Hipocampo/citologia , Técnicas In Vitro , Espaço Intracelular/genética , Masculino , Proteínas Mitocondriais/genética , Neurônios/fisiologia , Técnicas de Cultura de Órgãos , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , Ratos , Ratos Sprague-Dawley , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo , Proteínas rho de Ligação ao GTP/genética
9.
J Fluoresc ; 25(5): 1245-50, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26179074

RESUMO

Time-resolved microspectrofluorimetry and fluorescence microscopy imaging-two complementary fluorescence techniques-provide important information about the intracellular distribution, level of uptake and binding/interactions inside living cell of the labeled molecule of interest. They were employed to monitor the "fate" of AS1411 aptamer labeled by ATTO 425 in human living cells. Confocal microspectrofluorimeter adapted for time-resolved intracellular fluorescence measurements by using a phase-modulation principle with homodyne data acquisition was employed to obtain emission spectra and to determine fluorescence lifetimes in U-87 MG tumor brain cells and Hs68 non-tumor foreskin cells. Acquired spectra from both the intracellular space and the reference solutions were treated to observe the aptamer localization and its interaction with biological structures inside the living cell. The emission spectra and the maximum emission wavelengths coming from the cells are practically identical, however significant lifetime lengthening was observed for tumor cell line in comparison to non-tumor one.


Assuntos
Aptâmeros de Nucleotídeos/metabolismo , Espaço Intracelular/metabolismo , Microscopia de Fluorescência/métodos , Oligodesoxirribonucleotídeos/metabolismo , Espectrometria de Fluorescência/métodos , Sequência de Bases , Linhagem Celular Tumoral , Humanos , Espaço Intracelular/genética , Oligodesoxirribonucleotídeos/genética , Fatores de Tempo
10.
Virus Res ; 207: 136-45, 2015 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-25683509

RESUMO

The cellular prion protein (PrP(C)) is an ubiquitously expressed glycoprotein that is most abundant in the central nervous system. It is thought to play a role in many cellular processes, including neuroprotection, but may also contribute to Alzheimer's disease and some cancers. However, it is best known for its central role in the prion diseases, such as Creutzfeldt-Jakob disease (CJD), bovine spongiform encephalopathy (BSE), and scrapie. These protein misfolding diseases can be sporadic, acquired, or genetic and are caused by refolding of endogenous PrP(C) into a beta sheet-rich, pathogenic form, PrP(Sc). Once prions are present in the central nervous system, they increase and spread during a long incubation period that is followed by a relatively short clinical disease phase, ending in death. PrP molecules can be broadly categorized as either 'good' (cellular) PrP(C) or 'bad' (scrapie prion-type) PrP(Sc), but both populations are heterogeneous and different forms of PrP(C) may influence various cellular activities. Both PrP(C) and PrP(Sc) are localized predominantly at the cell surface, with the C-terminus attached to the plasma membrane via a glycosyl-phosphatidylinositol (GPI) anchor and both can exist in cleaved forms. PrP(C) also has cytosolic and transmembrane forms, and PrP(Sc) is known to exist in a variety of conformations and aggregation states. Here, we discuss the roles of different PrP isoforms in sickness and in health, and show the subcellular distributions of several forms of PrP that are particularly relevant for PrP(C) to PrP(Sc) conversion and prion-induced pathology in the hippocampus.


Assuntos
Espaço Intracelular/metabolismo , Proteínas PrPC/metabolismo , Proteínas PrPSc/metabolismo , Animais , Humanos , Espaço Intracelular/genética , Proteínas PrPC/genética , Proteínas PrPSc/genética , Transporte Proteico
11.
Neurology ; 82(22): 2007-16, 2014 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-24808017

RESUMO

OBJECTIVE: To identify a novel disease gene in 2 families with autosomal recessive hereditary spastic paraplegia (HSP). METHODS: We used whole-exome sequencing to identify the underlying genetic disease cause in 2 families with apparently autosomal recessive spastic paraplegia. Endogenous expression as well as subcellular localization of wild-type and mutant protein were studied to support the pathogenicity of the identified mutations. RESULTS: In 2 families, we identified compound heterozygous or homozygous mutations in the kinesin gene KIF1C to cause hereditary spastic paraplegia type 58 (SPG58). SPG58 can be complicated by cervical dystonia and cerebellar ataxia. The same mutations in a heterozygous state result in a mild or subclinical phenotype. KIF1C mutations in SPG58 affect the domains involved in adenosine triphosphate hydrolysis and microtubule binding, key functions for this microtubule-based motor protein. CONCLUSIONS: KIF1C is the third kinesin gene involved in the pathogenesis of HSPs and is characterized by a mild dominant and a more severe recessive disease phenotype. The identification of KIF1C as an HSP disease gene further supports the key role of intracellular trafficking processes in the pathogenesis of hereditary axonopathies.


Assuntos
Cinesinas/genética , Mutação/genética , Paraplegia Espástica Hereditária/genética , Adulto , Movimento Celular/genética , Feminino , Alemanha , Heterozigoto , Homozigoto , Humanos , Espaço Intracelular/genética , Masculino , Pessoa de Meia-Idade , Linhagem , Fenótipo , Índice de Gravidade de Doença
12.
PLoS One ; 8(8): e71032, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23936483

RESUMO

When canine adenovirus type 2 (CAdV-2, or also commonly referred to as CAV-2) vectors are injected into the brain parenchyma they preferentially transduce neurons, are capable of efficient axonal transport to afferent regions, and allow transgene expression for at last >1 yr. Yet, translating these data into a user-friendly vector platform has been limited because CAV-2 vector generation is challenging. Generation of E1-deleted adenovirus vectors often requires transfection of linear DNA fragments of >30 kb containing the vector genome into an E1-transcomplementing cell line. In contrast to human adenovirus type 5 vector generation, CAV-2 vector generation is less efficient due, in part, to a reduced ability to initiate replication and poor transfectibility of canine cells with large, linear DNA fragments. To improve CAV-2 vector generation, we generated an E1-transcomplementing cell line expressing the estrogen receptor (ER) fused to I-SceI, a yeast meganuclease, and plasmids containing the I-SceI recognition sites flanking the CAV-2 vector genome. Using transfection of supercoiled plasmid and intracellular genome release via 4-OH-tamoxifen-induced nuclear translocation of I-SceI, we improved CAV-2 vector titers 1,000 fold, and in turn increased the efficacy of CAV-2 vector generation.


Assuntos
Adenovirus Caninos/genética , Desoxirribonucleases de Sítio Específico do Tipo II/metabolismo , Vetores Genéticos/genética , Genoma/genética , Espaço Intracelular/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Transfecção/métodos , Animais , Linhagem Celular , Quebras de DNA de Cadeia Dupla , DNA Super-Helicoidal , Cães , Plasmídeos/genética , Receptores de Estrogênio/genética
13.
PLoS One ; 8(5): e62346, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23667468

RESUMO

The Ets transcription factor Fli-1 is preferentially expressed in hematopoietic tissues and cells, including immature T cells, but the role of Fli-1 in T cell development has not been closely examined. To address this we retrovirally overexpressed Fli-1 in various in vitro and in vivo settings and analysed its effect on T cell development. We found that Fli-1 overexpression perturbed the DN to DP transition and inhibited CD4 development whilst enhancing CD8 development both in vitro and in vivo. Surprisingly, Fli-1 overexpression in vivo eventuated in development of pre-T cell lymphoblastic leukaemia/lymphoma (pre-T LBL). Known Fli-1 target genes such as the pro-survival Bcl-2 family members were not found to be upregulated. In contrast, we found increased NOTCH1 expression in all Fli-1 T cells and detected Notch1 mutations in all tumours. These data show a novel function for Fli-1 in T cell development and leukaemogenesis and provide a new mouse model of pre-T LBL to identify treatment options that target the Fli-1 and Notch1 signalling pathways.


Assuntos
Carcinogênese/imunologia , Células-Tronco Hematopoéticas/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patologia , Proteína Proto-Oncogênica c-fli-1/genética , Linfócitos T/citologia , Linfócitos T/imunologia , Animais , Expressão Gênica , Humanos , Espaço Intracelular/genética , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Mensageiro/genética , Receptor Notch1/genética , Regulação para Cima/imunologia
14.
PLoS One ; 7(8): e43444, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22927968

RESUMO

Acid production from rhamnose is a characteristic phenotype of Listeria monocytogenes. We report the identification of the rhamnose transport and utilization operon located at lmo2846 to lmo2851, including the rhamnose-dependent promoter P(rha). Expression of reporter genes under control of P(rha) on a single copy integration vector demonstrated its suitability for inducible gene expression in L. monocytogenes. Transcription initiation from P(rha) is dose dependent, and a concentration as low as 100 µM rhamnose was found sufficient for induction. Moreover, P(rha) is subject to glucose catabolite repression, which provides additional options for strict control of expression. Infection of human THP1 macrophages revealed that P(rha) is repressed in intracellular L. monocytogenes, which is explained by the absence of rhamnose in the cytosol and possible interference by catabolite repression. The P(rha) promoter provides a novel and useful tool for triggering gene expression in extracellular L. monocytogenes, whereas intracellular conditions prevent transcription from this promoter.


Assuntos
Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Genes Bacterianos/genética , Listeria monocytogenes/efeitos dos fármacos , Listeria monocytogenes/genética , Ramnose/farmacologia , Linhagem Celular , Genes Reporter/genética , Vetores Genéticos/genética , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/genética , Listeria monocytogenes/citologia , Listeria monocytogenes/fisiologia , Macrófagos/fisiologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Transcrição Gênica/efeitos dos fármacos
15.
Endocr J ; 59(12): 1093-8, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22878668

RESUMO

Germline MEN1 mutation analysis is a powerful tool for an early diagnosis of multiple endocrine neoplasia type 1 (MEN1), an autosomal dominant familial cancer syndrome characterized by the parathyroid, pituitary and gastroenteropancreatic endocrine tumors. However, the clinical significance of MEN1 gene variants, especially missense and in-frame mutations as well as some splicing mutations, is not always obvious. We have previously shown that mutant menin proteins associated with MEN1 are rapidly degraded by the ubiquitin-proteasome pathway. We also demonstrated by a fluorescent immunocytochemical stability test that the stability of missense and in-frame deletion mutants varies widely but that unstable mutants were found only in MEN1 and related disorders and not in normal polymorphisms. In the present study, we evaluated by this stability test the pathogenicity of a novel MEN1 missense mutation, c.1118C>T, encoding a P373L mutant menin, identified in a suspected MEN1 patient. The results demonstrated that the mutant menin is highly unstable, indicating that this mutation is causative for MEN1. These findings encouraged us to proceed with presymptomatic genetic screening for this mutation among the family members, which resulted in the identification of asymptomatic mutation carriers. Thus, the information from the menin stability test was useful for genetic diagnosis and counseling of MEN1 in the case with a previously unreported MEN1 missense mutation.


Assuntos
Testes Genéticos/métodos , Neoplasia Endócrina Múltipla Tipo 1/diagnóstico , Mutação de Sentido Incorreto , Proteínas Proto-Oncogênicas/genética , Adulto , Idoso de 80 Anos ou mais , Análise Mutacional de DNA/métodos , Feminino , Instabilidade Genômica/genética , Humanos , Espaço Intracelular/genética , Masculino , Pessoa de Meia-Idade , Neoplasia Endócrina Múltipla Tipo 1/genética , Linhagem , Valor Preditivo dos Testes , Adulto Jovem
16.
Int J Cancer ; 130(3): 544-54, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21387291

RESUMO

Acheron (Achn) is a new member of the Lupus antigen family of RNA binding proteins. Previous studies have shown that Achn controls developmental decisions in neurons and muscle. In the human mammary gland, Achn expression is restricted to ductal myoepithelial cells. Microarray analysis and immunohistochemistry have shown that Achn expression is elevated in some basal-like ductal carcinomas. To study the possible role of Achn in breast cancer, we engineered human MDA-MB-231 cells to stably express enhanced green fluorescent protein-tagged wild-type Achn (AchnWT), as well as Achn lacking either its nuclear localization signal (AchnNLS) or its nuclear export signal (AchnNES). In in vitro assays, AchnWT and AchnNES, but not AchnNLS, enhanced cell proliferation, lamellipodia formation, and invasive activity and drove expression of the elevated expression of the metastasis-associated proteins MMP-9 and VEGF. To determine if Achn could alter the behavior of human breast cancer cells in vivo, Achn-engineered MDA-MB-231 cells were injected into athymic SCID/Beige mice. AchnWT and AchnNES-expressing tumors displayed enhanced angiogenesis and an approximately 5-fold increase in tumor size relative to either control cells or those expressing AchnNLS. These data suggest that Achn enhances human breast tumor growth and vascularization and that this activity is dependent on nuclear localization.


Assuntos
Autoantígenos/metabolismo , Neoplasias da Mama/etiologia , Ribonucleoproteínas/metabolismo , Animais , Autoantígenos/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Transformada , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Espaço Intracelular/genética , Espaço Intracelular/metabolismo , Camundongos , Camundongos SCID , Invasividade Neoplásica/genética , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteínas/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Antígeno SS-B
17.
PLoS One ; 7(12): e53023, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23285250

RESUMO

We evaluated transgenic tobacco plants as an alternative to Escherichia coli for the production of recombinant human complement factor 5a (C5a). C5a has not been expressed in plants before and is highly unstable in vivo in its native form, so it was necessary to establish the most suitable subcellular targeting strategy. We used the strong and constitutive CaMV 35S promoter to drive transgene expression and compared three different subcellular compartments. The yields of C5a in the T(0) transgenic plants were low in terms of the proportion of total soluble protein (TSP) when targeted to the apoplast (0.0002% TSP) or endoplasmic reticulum (0.0003% TSP) but was one order of magnitude higher when targeted to the vacuole (0.001% TSP). The yields could be increased by conventional breeding (up to 0.014% TSP in the T2 generation). C5a accumulated to the same level in seeds and leaves when targeted to the apoplast but was up to 1.7-fold more abundant in the seeds when targeted to the ER or vacuole, although this difference was less striking in the better-performing lines. When yields were calculated as an amount per gram fresh weight of transgenic plant tissue, the vacuole targeting strategy was clearly more efficient in seeds, reaching 35.8 µg C5a per gram of fresh seed weight compared to 10.62 µg C5a per gram fresh weight of leaves. Transient expression of C5aER and C5aVac in N. benthamiana, using MagnICON vectors, reached up to 0.2% and 0.7% of TSP, respectively, but was accompanied by cytotoxic effects and induced leaf senescence. Western blot of the plant extracts revealed a band matching the corresponding glycosylated native protein and the bioassay demonstrated that recombinant C5a was biologically active.


Assuntos
Complemento C5a/genética , Espaço Intracelular/metabolismo , Nicotiana/genética , Nicotiana/metabolismo , Proteínas Recombinantes/genética , Agrobacterium tumefaciens , Complemento C5a/metabolismo , Escherichia coli , Estudos de Viabilidade , Regulação da Expressão Gênica de Plantas/fisiologia , Marcação de Genes/métodos , Genes Reporter/genética , Humanos , Espaço Intracelular/genética , Plantas Geneticamente Modificadas , Proteínas Recombinantes/metabolismo , Especificidade da Espécie , Vírus do Mosaico do Tabaco/genética , Transformação Genética/fisiologia
18.
RNA ; 17(12): 2249-55, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22025736

RESUMO

Protein localization within cells can be achieved by the targeting and localized translation of mRNA. Yet, our understanding of the dynamics of mRNA targeting and protein localization, and of how general this phenomenon is, is not clear. Plasmid-based expression systems have been used to visualize exogenously expressed mRNAs and proteins; however, these methods typically produce them at levels greater than endogenous and can result in mislocalization. Hence, a method that allows for the simultaneous visualization of endogenous mRNAs and their translation products in living cells is needed. We previously developed a method (m-TAG) to localize endogenously expressed mRNAs in yeast by chromosomal insertion of the MS2 aptamer sequence between the open-reading frame (ORF) and 3' UTR of any gene. Upon coexpression with the MS2 RNA-binding coat protein (MS2-CP) fused with GFP, the aptamer-tagged mRNAs bearing their 3' UTRs are localized using fluorescence microscopy. Here we describe an advanced method (mp-TAG) that allows for the simultaneous visualization of both endogenously expressed mRNAs and their translation products in living yeast for the first time. Homologous recombination is used to insert the mCherry gene and MS2-CP binding sites downstream from any ORF, in order to localize protein and mRNA, respectively. As proof of the concept, we tagged ATP2 as a representative gene and demonstrated that endogenous ATP2 mRNA and protein localize to mitochondria, as shown previously. In addition, we demonstrate that tagged proteins like Hhf2, Vph1, and Yef3 localize to their expected subcellular location, while the localization of their mRNAs is revealed for the first time.


Assuntos
Proteínas Fúngicas/análise , Marcação de Genes/métodos , RNA Mensageiro/análise , Leveduras/genética , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Genoma Fúngico , Histonas/genética , Histonas/metabolismo , Recombinação Homóloga , Espaço Intracelular/genética , Espaço Intracelular/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Microscopia de Fluorescência , Mitocôndrias/genética , Mitocôndrias/metabolismo , Fatores de Alongamento de Peptídeos/genética , Fatores de Alongamento de Peptídeos/metabolismo , Biossíntese de Proteínas , Transporte Proteico , ATPases Translocadoras de Prótons/genética , ATPases Translocadoras de Prótons/metabolismo , Transporte de RNA , RNA Mensageiro/metabolismo , Leveduras/metabolismo
19.
J Neurosci Res ; 89(10): 1637-45, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21688291

RESUMO

Any mutations in the human neural cell adhesion molecule L1 (hL1CAM) gene might cause various types of serious neurological syndromes in humans, characterized by increased mortality, mental retardation, and various malformations of the nervous system. Such missense mutations often cause severe abnormalities or even fatalities, and the reason for this may be a disruption of the adhesive function of L1CAM resulting from a misdirection of the degradative pathway. Transfection studies using neuroblastoma N2a cells demonstrated that hL1CAM carrying the missense mutations in the fibronectin-like type III (FnIII) domains most likely is located within the endoplasmic reticulum (ER), but it is less well expressed on the cell surface. One mutant, L935P, in the fourth FnIII domain, was chosen from six mutants (K655 and G698 at Fn1, L935P and P941 at Fn4, W1036 and Y1070 at Fn5) in the FnIII domains to study in detail the functions of hL1CAM(200 kDa) , such as the intracellular traffic and degradation, because only a single band at 200 kDa was detected in the hL1CAM(L935P) -transfected cells. hL1CAM(200 kDa) is expressed predominantly in the ER but not on the cell surface. In addition, this missense mutated hL1CAM(200 kDa) is polyubiquitylated at some sites in the extracellular domain and thus becomes degraded by proteasomes via the ER-associated degradation pathway. These observations demonstrate that the missense mutations of hL1CAM in the FnIII domain may cause the resultant pathogenesis because of a loss of expression on the cell surface resulting from misrouting to the degradative pathway.


Assuntos
Retículo Endoplasmático/metabolismo , Fibronectinas/genética , Mutação de Sentido Incorreto/genética , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Neurônios/metabolismo , Ubiquitinação/genética , Animais , Linhagem Celular Tumoral , Retículo Endoplasmático/patologia , Fibronectinas/química , Fibronectinas/metabolismo , Fibronectinas/fisiologia , Humanos , Espaço Intracelular/genética , Espaço Intracelular/metabolismo , Espaço Intracelular/fisiologia , Camundongos , Mutagênese Sítio-Dirigida/métodos , Molécula L1 de Adesão de Célula Nervosa/química , Molécula L1 de Adesão de Célula Nervosa/genética , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Neurônios/patologia , Estrutura Terciária de Proteína/genética , Transporte Proteico/genética
20.
J Cell Sci ; 124(Pt 8): 1351-60, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21429935

RESUMO

SNAP25 regulates membrane fusion events at the plasma membrane and in the endosomal system, and a functional pool of the protein is delivered to recycling endosomes (REs) and the trans Golgi network (TGN) through an ARF6-dependent cycling pathway. SNAP25 is a peripheral membrane protein, and palmitoylation of a cluster of four cysteine residues mediates its stable association with the membrane. Here, we report that palmitoylation also determines the precise intracellular distribution of SNAP25, and that mutating single palmitoylation sites enhances the amount of SNAP25 at the RE and TGN. The farnesylated CAAX motif from Hras was ligated onto a SNAP25 mutant truncated immediately distal to the cysteine-rich domain. This construct displayed the same intracellular distribution as full-length SNAP25, and decreasing the number of cysteine residues in this construct increased its association with the RE and TGN, confirming the dominant role of the cysteine-rich domain in directing the intracellular distribution of SNAP25. Marked differences in the localisations of SNAP25-CAAX and Hras constructs, each with two palmitoylation sites, were observed, showing that subtle differences in palmitoylated sequences can have a major impact upon intracellular targeting. We propose that the cysteine-rich domain of SNAP25 is designed to facilitate the dual function of this SNARE protein at the plasma membrane and endosomes, and that dynamic palmitoylation acts as a mechanism to regulate the precise intracellular patterning of SNAP25.


Assuntos
Espaço Intracelular/metabolismo , Proteína 25 Associada a Sinaptossoma/metabolismo , Animais , Membrana Celular/genética , Membrana Celular/metabolismo , Endossomos/genética , Endossomos/metabolismo , Espaço Intracelular/genética , Lipoilação , Células PC12 , Transporte Proteico , Ratos , Proteína 25 Associada a Sinaptossoma/genética , Rede trans-Golgi/genética , Rede trans-Golgi/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA