Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.064
Filtrar
Mais filtros

Eixos temáticos
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(3)2024 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-38339168

RESUMO

Differentiation-inducing factor 1 (DIF-1), found in Dictyostelium discoideum, has antiproliferative and glucose-uptake-promoting activities in mammalian cells. DIF-1 is a potential lead for the development of antitumor and/or antiobesity/antidiabetes drugs, but the mechanisms underlying its actions have not been fully elucidated. In this study, we searched for target molecules of DIF-1 that mediate the actions of DIF-1 in mammalian cells by identifying DIF-1-binding proteins in human cervical cancer HeLa cells and mouse 3T3-L1 fibroblast cells using affinity chromatography and liquid chromatography-tandem mass spectrometry and found mitochondrial malate dehydrogenase (MDH2) to be a DIF-1-binding protein in both cell lines. Since DIF-1 has been shown to directly inhibit MDH2 activity, we compared the effects of DIF-1 and the MDH2 inhibitor LW6 on the growth of HeLa and 3T3-L1 cells and on glucose uptake in confluent 3T3-L1 cells in vitro. In both HeLa and 3T3-L1 cells, DIF-1 at 10-40 µM dose-dependently suppressed growth, whereas LW6 at 20 µM, but not at 2-10 µM, significantly suppressed growth in these cells. In confluent 3T3-L1 cells, DIF-1 at 10-40 µM significantly promoted glucose uptake, with the strongest effect at 20 µM DIF-1, whereas LW6 at 2-20 µM significantly promoted glucose uptake, with the strongest effect at 10 µM LW6. Western blot analyses showed that LW6 (10 µM) and DIF-1 (20 µM) phosphorylated and, thus, activated AMP kinase in 3T3-L1 cells. Our results suggest that MDH2 inhibition can suppress cell growth and promote glucose uptake in the cells, but appears to promote glucose uptake more strongly than it suppresses cell growth. Thus, DIF-1 may promote glucose uptake, at least in part, via direct inhibition of MDH2 and a subsequent activation of AMP kinase in 3T3-L1 cells.


Assuntos
Glucose , Malato Desidrogenase , Animais , Humanos , Camundongos , Células 3T3-L1/efeitos dos fármacos , Células 3T3-L1/metabolismo , Adenilato Quinase/metabolismo , Dictyostelium/metabolismo , Glucose/metabolismo , Células HeLa/efeitos dos fármacos , Células HeLa/metabolismo , Malato Desidrogenase/antagonistas & inibidores , Malato Desidrogenase/metabolismo , Mamíferos/metabolismo
2.
Proc Natl Acad Sci U S A ; 116(33): 16479-16488, 2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31346090

RESUMO

Regulation of IFN signaling is critical in host recognition and response to pathogens while its dysregulation underlies the pathogenesis of several chronic diseases. STimulator of IFN Genes (STING) has been identified as a critical mediator of IFN inducing innate immune pathways, but little is known about direct coregulators of this protein. We report here that TMEM203, a conserved putative transmembrane protein, is an intracellular regulator of STING-mediated signaling. We show that TMEM203 interacts, functionally cooperates, and comigrates with STING following cell stimulation, which in turn leads to the activation of the kinase TBK1, and the IRF3 transcription factor. This induces target genes in macrophages, including IFN-ß. Using Tmem203 knockout bone marrow-derived macrophages and transient knockdown of TMEM203 in human monocyte-derived macrophages, we show that TMEM203 protein is required for cGAMP-induced STING activation. Unlike STING, TMEM203 mRNA levels are elevated in T cells from patients with systemic lupus erythematosus, a disease characterized by the overexpression of type I interferons. Moreover, TMEM203 mRNA levels are associated with disease activity, as assessed by serum levels of the complement protein C3. Identification of TMEM203 sheds light into the control of STING-mediated innate immune responses, providing a potential novel mechanism for therapeutic interventions in STING-associated inflammatory diseases.


Assuntos
Inflamação/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Proteínas de Membrana/metabolismo , Transdução de Sinais , Sequência Conservada , Regulação para Baixo , Evolução Molecular , Células HeLa/metabolismo , Humanos , Inflamação/patologia , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/metabolismo , Lúpus Eritematoso Sistêmico/metabolismo , Lúpus Eritematoso Sistêmico/patologia , Lisossomos/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/genética , Nucleotídeos Cíclicos/metabolismo , Ligação Proteica , Domínios Proteicos , Proteínas Serina-Treonina Quinases/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Molécula 1 de Interação Estromal/metabolismo
3.
Mol Pharm ; 18(10): 3750-3762, 2021 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-34491767

RESUMO

Arg-Arg-Leu (RRL) is a potent tumor-homing tripeptide. However, the binding target is unclear. In this study, we intended to identify the binding target of RRL and evaluate the tumor targeting of 99mTc-MAG3-RRL in vivo. Biotin-RRL, 5-TAMRA-RRL, and 99mTc-MAG3-RRL were designed to trace the binding target and tumor lesion. Immunoprecipitation-mass spectrometry was conducted to identify the candidate proteins and determination of the subcellular localization was also performed. A pull-down assay was performed to demonstrate the immunoprecipitate. Fluorescence colocalization and cell uptake assays were performed to elucidate the correlation between the selected binding protein and RRL, and the internalization mechanism of RRL. Biodistribution and in vivo imaging were performed to evaluate the tumor accumulation and targeting of 99mTc-MAG3-RRL. The target for RRL was screened to be heat shock protein 70 (HSP70). The prominent uptake distribution of RRL was concentrated in the membrane and cytoplasm. A pull-down assay demonstrated the existence of HSP70 in the biotin-RRL captured complex. Regarding fluorescence colocalization and cell uptake assays, RRL may interact with HSP70 at the nucleotide-binding domain (NBD). Clathrin-dependent endocytosis and macropinocytosis could be a vital internalization mechanism of RRL. In vivo imaging and biodistribution both demonstrated that 99mTc-MAG3-RRL can trace tumors with satisfactory accumulation in hepatoma xenograft mice. The radioactive signals accumulated in tumor lesions can be blocked by VER-155008, which can bind to the NBD of HSP70. Our findings revealed that RRL may interact with HSP70 and that 99mTc-MAG3-RRL could be a prospective probe for visualizing overexpressed HSP70 tumor sections.


Assuntos
Proteínas de Choque Térmico HSP70/metabolismo , Neoplasias/diagnóstico por imagem , Oligopeptídeos/metabolismo , Animais , Sítios de Ligação , Feminino , Citometria de Fluxo , Células HeLa/metabolismo , Células Hep G2/metabolismo , Humanos , Imunoprecipitação , Células MCF-7/metabolismo , Espectrometria de Massas , Camundongos , Camundongos Nus , Imagem Óptica , Tomografia Computadorizada de Emissão de Fóton Único
4.
Infect Immun ; 88(3)2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-31843963

RESUMO

Salmonella enterica serovar Pullorum is the pathogen of pullorum disease, which leads to severe economic losses in many developing countries. In contrast to the strong inflammatory response induced by Salmonella enterica serovar Typhimurium and Salmonella enterica serovar Enteritidis, S Pullorum causes systemic infection with little inflammation. The effector proteins secreted by Salmonella often play a crucial role in modulating host signal transduction and cellular processes to the pathogen's advantage. In the present study, the invasion plasmid antigen J (IpaJ) protein specifically identified in S Pullorum was found to significantly inhibit activation of the key proinflammatory transcription factor, NF-κB, which was induced by tumor necrosis factor alpha (TNF-α), interleukin-1ß (IL-1ß), and lipopolysaccharide (LPS). IpaJ inhibited the NF-κB pathway in cells infected with S Pullorum through the stabilization of IκBα. Deletion of ipaJ in S Pullorum caused a significantly increased level of ubiquitinated IκBα that was subsequently degraded by the proteasome in HeLa cells. Moreover, IpaJ was efficient in the prevention of NF-κB translocation to the nucleus and ultimately interfered with the secretion of the proinflammatory cytokines IL-1ß, IL-6, and IL-8 in infected HeLa cells. Additionally, the transformation of ipaJ into S Enteritidis decreased the secretion of proinflammatory cytokines in HeLa cells through suppression of the NF-κB pathway. The infection of chicken peripheral blood monocyte-derived macrophages (chMDM) confirmed that ipaJ-deleted S Pullorum induced a stronger expression of proinflammatory cytokines than the wild-type and complementary strains. In summary, the present study revealed that IpaJ functions as an important anti-inflammatory protein involved in S Pullorum infection through inhibition of the NF-κB pathway and the subsequent inflammatory response.


Assuntos
Antígenos de Bactérias/imunologia , NF-kappa B/imunologia , Salmonelose Animal/imunologia , Salmonella enterica/patogenicidade , Ubiquitinação/fisiologia , Animais , Galinhas , Células HeLa/metabolismo , Humanos , Interleucinas/metabolismo
5.
EMBO J ; 35(8): 866-80, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26988033

RESUMO

Autophagy, mediated by a number of autophagy-related (ATG) proteins, plays an important role in the bulk degradation of cellular constituents. Beclin-1 (also known as Atg6 in yeast) is a core protein essential for autophagic initiation and other biological processes. The activity of Beclin-1 is tightly regulated by multiple post-translational modifications, including ubiquitination, yet the molecular mechanism underpinning its reversible deubiquitination remains poorly defined. Here, we identified ubiquitin-specific protease 19 (USP19) as a positive regulator of autophagy, but a negative regulator of type I interferon (IFN) signaling.USP19 stabilizes Beclin-1 by removing the K11-linked ubiquitin chains of Beclin-1 at lysine 437. Moreover, we foundthat USP19 negatively regulates type IIFNsignaling pathway, by blockingRIG-I-MAVSinteraction in a Beclin-1-dependent manner. Depletion of eitherUSP19 or Beclin-1 inhibits autophagic flux and promotes type IIFNsignaling as well as cellular antiviral immunity. Our findings reveal novel dual functions of theUSP19-Beclin-1 axis by balancing autophagy and the production of type IIFNs.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Autofagia/fisiologia , Endopeptidases/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/imunologia , Proteína Beclina-1 , Linhagem Celular/virologia , Proteína DEAD-box 58 , RNA Helicases DEAD-box/metabolismo , Endopeptidases/genética , Endopeptidases/imunologia , Células HeLa/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Vírus da Influenza A/patogenicidade , Interferon Tipo I/metabolismo , Lisina/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Estabilidade Proteica , Receptores Imunológicos , Transdução de Sinais/fisiologia , Ubiquitinação
6.
Med Sci Monit ; 26: e919757, 2020 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-31894756

RESUMO

BACKGROUND S100 calcium-binding protein A16 (S100A16) is closely related to the onset and progression of tumors. MATERIAL AND METHODS In the research, the mainly purpose was to investigate the effect of S100A16 on the proliferation ability, invasion, and angiogenesis of HeLa cells. An adenoviral vector overexpressing S100A16 (Ad-S100A16) was constructed and transfected into HeLa cells, forming a stable cells line of overexpression. The effect of S100A16 on the proliferative capacity of HeLa cells was evaluated by a Cell Counting Kit-8 (CCK-8) assay. Cell migration capacity was determined by a Transwell migration assay. Changes in matrix metalloproteinase-2 (MMP-2), MMP-9, E-cadherin, and vimentin expression were evaluated by a cell-based immunofluorescence assay. The effect of S100A16 on angiogenesis was verified by knockout experiment. RESULTS Overexpression of S100A16 significantly enhanced the proliferative and migratory capacities of HeLa cells (P<0.05), upregulated expression of matrix MMP-2, MMP-9, vimentin, phosphatidylinositol 3 kinase, and phosphorylated protein kinase B, and downregulated expression of E-cadherin. Vascular endothelial growth factor expression increased, phosphatase and tensin homolog expression decreased, and angiogenesis was positively correlated with S100A16 expression. These effects were largely mediated by the activation of the phosphatidylinositol 3 kinase/protein kinase B pathways. CONCLUSIONS S100A16 could promote the proliferation, migration, and tumor angiogenesis of HeLa cells by regulating the phosphatidylinositol 3 kinase/protein kinase B signaling pathways.


Assuntos
Células HeLa/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas S100/metabolismo , Antígenos CD/metabolismo , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Invasividade Neoplásica , Neovascularização Patológica/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Vimentina/metabolismo
7.
Hum Mol Genet ; 26(9): 1622-1633, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28334782

RESUMO

Numerous GJB1 gene mutations cause the X-linked form of Charcot-Marie-Tooth disease (CMT1X). GJB1 encodes connexin32 (Cx32), which forms trans-myelin gap junctions in Schwann cells. Most GJB1 mutations result in loss-of-function mechanisms, supporting the concept of gene replacement therapy. However, interactions between delivered wild type and endogenously expressed mutant Cx32 may potentially occur in the setting of gene replacement therapy. In order to screen for possible interactions of several representative CMT1X mutants with wild type Cx32 that may interfere with the functional gap junction formation, we established an in vitro screening method co-expressing in HeLa cells wild type Cx32 and one of eight different Cx32 mutants including A39P, A39V, T55I, R75W, M93V, L143P, N175D and R183S. Some of the Golgi-retained mutants hindered gap junction plaque assembly by Cx32 on the cell membrane, while co-immunoprecipitation analysis revealed a partial interaction of wild type protein with Golgi-retained mutants. Dye transfer studies confirmed that Golgi-retained R75W, M93V and N175D but not endoplasmic reticulum-retained T55I had a negative effect on wild type Cx32 function. Finally, in vivo intraneural delivery of the gene encoding the wild type Cx32 in mice bearing either the T55I or R75W mutation on Cx32 knockout background showed that virally delivered protein was correctly localized in mice expressing the endoplasmic reticulum-retained T55I whereas it did not traffic normally in mice expressing the Golgi-retained R75W. Thus, certain Golgi-retained Cx32 mutants may interfere with exogenously delivered Cx32. Screening for mutant-wild type Cx32 interactions should be considered prior to planning gene addition therapy for CMT1X.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Conexinas/genética , Conexinas/metabolismo , Animais , Doença de Charcot-Marie-Tooth/metabolismo , Retículo Endoplasmático/metabolismo , Junções Comunicantes/metabolismo , Terapia Genética/métodos , Complexo de Golgi/genética , Complexo de Golgi/fisiologia , Células HeLa/metabolismo , Humanos , Camundongos , Camundongos Knockout , Mutação , Bainha de Mielina/metabolismo , Células de Schwann/metabolismo , Proteína beta-1 de Junções Comunicantes
8.
Genes Cells ; 23(12): 1023-1042, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30318703

RESUMO

The centrosome is a small but important organelle that participates in centriole duplication, spindle formation, and ciliogenesis. Each event is regulated by key enzymatic reactions, but how these processes are integrated remains unknown. Recent studies have reported that ciliogenesis is controlled by distal appendage proteins such as FBF1, also known as Albatross. However, the precise role of Albatross in the centrosome cycle, including centriole duplication and centrosome separation, remains to be determined. Here, we report a novel function for Albatross at the proximal ends of centrioles. Using Albatross monospecific antibodies, full-length constructs, and siRNAs for rescue experiments, we found that Albatross mediates centriole duplication by recruiting HsSAS-6, a cartwheel protein of centrioles. Moreover, Albatross participates in centrosome separation during mitosis by recruiting Plk1 to residue S348 of Albatross after its phosphorylation. Taken together, our results show that Albatross is a novel protein that spatiotemporally integrates different aspects of centrosome function, namely ciliogenesis, centriole duplication, and centrosome separation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Centríolos/metabolismo , Centrossomo/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Animais , Proteínas de Ciclo Celular/metabolismo , Células HEK293 , Células HeLa/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Fosforilação , Fosfosserina/metabolismo , Ligação Proteica , Domínios Proteicos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Serina/metabolismo , Quinase 1 Polo-Like
9.
Brain ; 141(5): 1286-1299, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29481671

RESUMO

Many genetic neurological disorders exhibit variable expression within affected families, often exemplified by variations in disease age at onset. Epistatic effects (i.e. effects of modifier genes on the disease gene) may underlie this variation, but the mechanistic basis for such epistatic interactions is rarely understood. Here we report a novel epistatic interaction between SPAST and the contiguous gene DPY30, which modifies age at onset in hereditary spastic paraplegia, a genetic axonopathy. We found that patients with hereditary spastic paraplegia caused by genomic deletions of SPAST that extended into DPY30 had a significantly younger age at onset. We show that, like spastin, the protein encoded by SPAST, the DPY30 protein controls endosomal tubule fission, traffic of mannose 6-phosphate receptors from endosomes to the Golgi, and lysosomal ultrastructural morphology. We propose that additive effects on this pathway explain the reduced age at onset of hereditary spastic paraplegia in patients who are haploinsufficient for both genes.


Assuntos
Epistasia Genética/genética , Mutação/genética , Proteínas Nucleares/genética , Paraplegia Espástica Hereditária/genética , Espastina/genética , Adulto , Idade de Início , Antígenos CD8/genética , Antígenos CD8/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Células HeLa/metabolismo , Células HeLa/ultraestrutura , Humanos , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Proteína 1 de Membrana Associada ao Lisossomo/ultraestrutura , Lisossomos/metabolismo , Lisossomos/ultraestrutura , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Proteínas Nucleares/metabolismo , Proteínas Nucleares/ultraestrutura , Transporte Proteico/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
10.
Med Sci Monit ; 25: 3617-3623, 2019 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-31092810

RESUMO

BACKGROUND Senescence is a natural barrier for the body to resist the malignant transformation of its own cells. This work investigated the senescence characteristics of cancer cells in vitro. MATERIAL AND METHODS Human cervical cancer HeLa cells were treated with different concentrations of doxorubicin for 3 days, with or without subsequent extended culture in drug-free medium for 6 days. Senescent cell ratios between these 2 culture schemes were calculated. Expression of 2 senescence-associated secretory factors, IL-6 and IL-8, were detected by RT-PCR and ELISA. Doxorubicin treatment induced epithelial-mesenchymal transition in cancer cells. The proportions of senescent cells in epithelial-like and mesenchymal-like sub-groups were calculated. Doxorubicin-treated HeLa cells were stained with Vimentin antibody and sorted by flow cytometry. Senescent cell marker p16ᴵᴺᴷ4ᵃ and IL-8 expression in Vimentin-high and Vimentin-low cells were detected by Western blot. RESULTS We found that less than 1% of HeLa cells showed senescence phenotype after treatment with doxorubicin for 3 days. However, the proportion of senescent cells was significantly increased when the doxorubicin-treated cells were subsequently cultured in drug-free medium for another 6d. RT-PCR and ELISA results showed that this prolonged culture method could further improve the expression of IL-6 and IL-8. We also found that the senescent cells were mainly epithelial-like type and few presented mesenchymal-like shape. p16ᴵᴺᴷ4ᵃ and IL-8 expression were decreased in cell fraction with higher Vimentin expression. CONCLUSIONS Our results suggested the existence of time delay effect in doxorubicin-induced senescence of HeLa cells, and epithelial- mesenchymal transition may resist doxorubicin-induced cell senescence.


Assuntos
Senescência Celular/efeitos dos fármacos , Doxorrubicina/farmacologia , Neoplasias/metabolismo , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transição Epitelial-Mesenquimal/genética , Células HeLa/metabolismo , Humanos , Interleucina-6/análise , Interleucina-8/análise , Fatores de Tempo , Vimentina/metabolismo
11.
Chaos ; 29(3): 033132, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30927859

RESUMO

Previous experiments demonstrated that a population of HeLa cells starved of glucose or both glucose and serum exhibited a strong heterogeneity in the glycolytic oscillations in terms of the number of oscillatory cells, periods of oscillations, and duration of oscillations. Here, we report numerical simulations of this heterogeneous oscillatory behavior in HeLa cells by using a newly developed mathematical model. It is simple enough that we can apply a mathematical analysis, but capture the core of the glycolytic pathway and the activity of the glucose transporter (GLUT). Lognormal distributions of the values of the four rate constants in the model were obtained from the experimental distributions in the periods of oscillations. Thus, the heterogeneity in the periods of oscillations can be attributed to the difference in the rate constants of the enzymatic reactions. The activity of GLUT is found to determine whether the HeLa cells were oscillatory or non-oscillatory under the same experimental conditions. Simulation with the log-normal distribution of the maximum uptake velocity of glucose and the four randomized rate constants based on the log-normal distributions successfully reproduced the time-dependent number of oscillatory cells (oscillatory ratios) under the two starving conditions. The difference in the initial values of the metabolites has little effect on the simulated results.


Assuntos
Glicólise , Células HeLa/metabolismo , Neoplasias do Colo do Útero/enzimologia , Fenômenos Fisiológicos Celulares , Feminino , Humanos , Modelos Biológicos
12.
J Appl Toxicol ; 38(5): 734-743, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29350772

RESUMO

It is evident that trivalent arsenicals do not have mutagenicity, but they are human carcinogens. Recently, epigenetic modification has been considered as one of the important causes of arsenical carcinogenicity. Here we examined global histone H3 modification by trivalent inorganic arsenite (iAs(III)) and its contribution to gene expression in HeLa cells. iAs(III) induced histone H3K9 dimethylation (H3K9me2) and trimethylation (H3K9me3), histone H3S10 phosphorylation (H3S10p), histone H3T11 phosphorylation (H3T11p) and histone H3K9S10 trimethyl-phosphorylation (H3K9me3S10p). Among these modifications, H3S10p, H3T11p and H3K9me3S10p were observed as a punctate signal in interphase cells, which seems to associate with remodeling of the chromatin structure at the specific locus. A chromatin immunoprecipitation assay was performed to examine histone H3 modifications around the FOS, EGR1 and IL8 promoters, as previous studies revealed some relation between histone H3 modification and induction of these genes. iAs(III) increased H3S10p and H3K9me3S10p in the FOS promoter around the SRE/ELK1 binding site (-400 to -200) and CRE-binding site (-50). In contrast, histone H3 around the EGR1 promoter of SRE/CRE-binding site (-200 to -50) was modified to H3S10p and H3K9me3S10p by iAs(III). Reporter gene assays with deletion mutants of the FOS and EGR1 promoters revealed that the around SRE/ELK1 site is important for iAs(III)-mediated FOS induction, and SRE/CRE site for EGR1 induction. Collectively, these results demonstrate that iAs(III) induces histone H3 modifications around the transcription factor binding sites of the FOS and EGR1 promoter, and these modifications seem to be important in transcriptional activation of these genes.


Assuntos
Arsenitos/toxicidade , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Células HeLa/efeitos dos fármacos , Histonas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Imunoprecipitação da Cromatina , Imunofluorescência , Células HeLa/metabolismo , Histonas/metabolismo , Humanos
13.
Proc Natl Acad Sci U S A ; 112(33): 10154-61, 2015 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-26195779

RESUMO

Mitochondria are energy-producing organelles in eukaryotic cells considered to be of bacterial origin. The mitochondrial genome has evolved under selection for minimization of gene content, yet it is not known why not all mitochondrial genes have been transferred to the nuclear genome. Here, we predict that hydrophobic membrane proteins encoded by the mitochondrial genomes would be recognized by the signal recognition particle and targeted to the endoplasmic reticulum if they were nuclear-encoded and translated in the cytoplasm. Expression of the mitochondrially encoded proteins Cytochrome oxidase subunit 1, Apocytochrome b, and ATP synthase subunit 6 in the cytoplasm of HeLa cells confirms export to the endoplasmic reticulum. To examine the extent to which the mitochondrial proteome is driven by selective constraints within the eukaryotic cell, we investigated the occurrence of mitochondrial protein domains in bacteria and eukaryotes. The accessory protein domains of the oxidative phosphorylation system are unique to mitochondria, indicating the evolution of new protein folds. Most of the identified domains in the accessory proteins of the ribosome are also found in eukaryotic proteins of other functions and locations. Overall, one-third of the protein domains identified in mitochondrial proteins are only rarely found in bacteria. We conclude that the mitochondrial genome has been maintained to ensure the correct localization of highly hydrophobic membrane proteins. Taken together, the results suggest that selective constraints on the eukaryotic cell have played a major role in modulating the evolution of the mitochondrial genome and proteome.


Assuntos
Genoma Mitocondrial/genética , Proteínas Mitocondriais/metabolismo , Proteínas de Bactérias/metabolismo , Núcleo Celular/genética , Proteínas de Cloroplastos/metabolismo , Biologia Computacional , Citocromos b/metabolismo , Citosol/metabolismo , Retículo Endoplasmático/metabolismo , Células HeLa/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Proteínas de Membrana/genética , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Fosforilação Oxidativa , Filogenia , Dobramento de Proteína , Estrutura Terciária de Proteína , Partícula de Reconhecimento de Sinal/metabolismo , Termodinâmica
14.
EMBO J ; 32(24): 3220-30, 2013 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-24213245

RESUMO

Nuclear pore complexes (NPCs) mediate cargo traffic between the nucleus and the cytoplasm of eukaryotic cells. Nuclear transport receptors (NTRs) carry cargos through NPCs by transiently binding to phenylalanine-glycine (FG) repeats on intrinsically disordered polypeptides decorating the NPCs. Major impediments to understand the transport mechanism are the thousands of FG binding sites on each NPC, whose spatial distribution is unknown, and multiple binding sites per NTR, which leads to multivalent interactions. Using single molecule fluorescence microscopy, we show that multiple NTR molecules are required for efficient transport of a large cargo, while a single NTR promotes binding to the NPC but not transport. Particle trajectories and theoretical modelling reveal a crucial role for multivalent NTR interactions with the FG network and indicate a non-uniform FG repeat distribution. A quantitative model is developed wherein the cytoplasmic side of the pore is characterized by a low effective concentration of free FG repeats and a weak FG-NTR affinity, and the centrally located dense permeability barrier is overcome by multivalent interactions, which provide the affinity necessary to permeate the barrier.


Assuntos
Modelos Moleculares , Complexo de Proteínas Formadoras de Poros Nucleares/química , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Poro Nuclear/metabolismo , Transporte Ativo do Núcleo Celular , Glicina/química , Glicina/metabolismo , Células HeLa/metabolismo , Humanos , Carioferinas/metabolismo , Permeabilidade , Fenilalanina/química , Fenilalanina/metabolismo , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
15.
Cell Biol Int ; 41(10): 1103-1109, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28675500

RESUMO

FAM107B expression was decreased in stomach cancer and many other kinds of cancer. The forced expression of FAM107B in HeLa cells diminished proliferation in response to growth factors, suggesting that FAM107B might play important roles in many types of cancers. But the mechanisms underlying the decreased expression of FAM107B in cancers are not clear, the functional significance needs to be further clarified. Our previous findings from cDNA microarray showed that there are 179 differentially expressed genes after S100A4 inhibition in gastric cancer cells MGC803. FAM107B was an upregulated one among them. In the present study, we confirmed that FAM107B expression was upregulated in MGC803 cells after S100A4 inhibition by qRT-PCR. We demonstrated for the first time that FAM107B was downregulated by S100A4. The results from CCK-8 and transwell assay showed that FAM107B inhibition by siRNA led to significantly increased proliferation and migrating abilities of MGC803 cells, respectively, indicating that FAM107B plays important roles in inhibiting the proliferation and migration of MGC803 cells. The rescue experiment showed that FAM107B-siRNA transfection reversed the reduced proliferation and migration abilities induced by S100A4 inhibition in the cells. These findings suggest that, as a downstream effector, FAM107B at least partly mediates the effect of S100A4 on the proliferation and migration of MGC803 cells. In conclusion, we first provide experimental evidence suggesting that FAM107B was downregulated by S100A4 in gastric cancer MGC803 cells. And FAM107B at least partially mediates the biological effect of S100A4 in the cells.


Assuntos
Proteínas Nucleares/biossíntese , Proteína A4 de Ligação a Cálcio da Família S100/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Apoptose/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Regulação para Baixo , Genes Supressores de Tumor , Células HeLa/metabolismo , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Proteína A4 de Ligação a Cálcio da Família S100/antagonistas & inibidores , Proteína A4 de Ligação a Cálcio da Família S100/metabolismo , Transdução de Sinais , Neoplasias Gástricas/metabolismo , Transfecção
16.
PLoS Genet ; 10(10): e1004626, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25275521

RESUMO

Deficiency of autophagy protein beclin 1 is implicated in tumorigenesis and neurodegenerative diseases, but the molecular mechanism remains elusive. Previous studies showed that Beclin 1 coordinates the assembly of multiple VPS34 complexes whose distinct phosphatidylinositol 3-kinase III (PI3K-III) lipid kinase activities regulate autophagy at different steps. Recent evidence suggests a function of beclin 1 in regulating multiple VPS34-mediated trafficking pathways beyond autophagy; however, the precise role of beclin 1 in autophagy-independent cellular functions remains poorly understood. Herein we report that beclin 1 regulates endocytosis, in addition to autophagy, and is required for neuron viability in vivo. We find that neuronal beclin 1 associates with endosomes and regulates EEA1/early endosome localization and late endosome formation. Beclin 1 maintains proper cellular phosphatidylinositol 3-phosphate (PI(3)P) distribution and total levels, and loss of beclin 1 causes a disruption of active Rab5 GTPase-associated endosome formation and impairment of endosome maturation, likely due to a failure of Rab5 to recruit VPS34. Furthermore, we find that Beclin 1 deficiency causes complete loss of the UVRAG-VPS34 complex and associated lipid kinase activity. Interestingly, beclin 1 deficiency impairs p40phox-linked endosome formation, which is rescued by overexpressed UVRAG or beclin 1, but not by a coiled-coil domain-truncated beclin 1 (a UVRAG-binding mutant), Atg14L or RUBICON. Thus, our study reveals the essential role for beclin 1 in neuron survival involving multiple membrane trafficking pathways including endocytosis and autophagy, and suggests that the UVRAG-beclin 1 interaction underlies beclin 1's function in endocytosis.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Classe III de Fosfatidilinositol 3-Quinases/metabolismo , Endossomos/metabolismo , Neurônios/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Proteínas Reguladoras de Apoptose/genética , Autofagia/genética , Proteína Beclina-1 , Classe III de Fosfatidilinositol 3-Quinases/genética , Endocitose/genética , Receptores ErbB/metabolismo , Células HeLa/metabolismo , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neurônios/patologia , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas rab5 de Ligação ao GTP/metabolismo
17.
EMBO J ; 31(2): 301-16, 2012 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-22068056

RESUMO

Tissue-specific transcriptional activators initiate differentiation towards specialized cell types by inducing chromatin modifications permissive for transcription at target loci, through the recruitment of SWItch/Sucrose NonFermentable (SWI/SNF) chromatin-remodelling complex. However, the molecular mechanism that regulates SWI/SNF nuclear distribution in response to differentiation signals is unknown. We show that the muscle determination factor MyoD and the SWI/SNF subunit BAF60c interact on the regulatory elements of MyoD-target genes in myoblasts, prior to activation of transcription. BAF60c facilitates MyoD binding to target genes and marks the chromatin for signal-dependent recruitment of the SWI/SNF core to muscle genes. BAF60c phosphorylation on a conserved threonine by differentiation-activated p38α kinase is the signal that promotes incorporation of MyoD-BAF60c into a Brg1-based SWI/SNF complex, which remodels the chromatin and activates transcription of MyoD-target genes. Our data support an unprecedented two-step model by which pre-assembled BAF60c-MyoD complex directs recruitment of SWI/SNF to muscle loci in response to differentiation cues.


Assuntos
Proteínas Cromossômicas não Histona/fisiologia , Sistema de Sinalização das MAP Quinases , Desenvolvimento Muscular/fisiologia , Proteínas Musculares/fisiologia , Proteína MyoD/fisiologia , Fatores de Transcrição/fisiologia , Animais , Linhagem Celular , Cromatina/genética , Proteínas Cromossômicas não Histona/antagonistas & inibidores , Proteínas Cromossômicas não Histona/química , Proteínas Cromossômicas não Histona/genética , DNA Helicases/fisiologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica/genética , Células HeLa/metabolismo , Humanos , Camundongos , Complexos Multiproteicos , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/química , Proteínas Musculares/genética , Mioblastos/metabolismo , Proteínas Nucleares/fisiologia , Fosforilação , Fosfotreonina/análise , Processamento de Proteína Pós-Traducional , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/química , Fatores de Transcrição/genética , Técnicas do Sistema de Duplo-Híbrido , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
18.
Biophys J ; 109(2): 182-93, 2015 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-26200855

RESUMO

The heat-shock response is a key factor in diverse stress scenarios, ranging from hyperthermia to protein folding diseases. However, the complex dynamics of this physiological response have eluded mathematical modeling efforts. Although several computational models have attempted to characterize the heat-shock response, they were unable to model its dynamics across diverse experimental datasets. To address this limitation, we mined the literature to obtain a compendium of in vitro hyperthermia experiments investigating the heat-shock response in HeLa cells. We identified mechanisms previously discussed in the experimental literature, such as temperature-dependent transcription, translation, and heat-shock factor (HSF) oligomerization, as well as the role of heat-shock protein mRNA, and constructed an expanded mathematical model to explain the temperature-varying DNA-binding dynamics, the presence of free HSF during homeostasis and the initial phase of the heat-shock response, and heat-shock protein dynamics in the long-term heat-shock response. In addition, our model was able to consistently predict the extent of damage produced by different combinations of exposure temperatures and durations, which were validated against known cellular-response patterns. Our model was also in agreement with experiments showing that the number of HSF molecules in a HeLa cell is roughly 100 times greater than the number of stress-activated heat-shock element sites, further confirming the model's ability to reproduce experimental results not used in model calibration. Finally, a sensitivity analysis revealed that altering the homeostatic concentration of HSF can lead to large changes in the stress response without significantly impacting the homeostatic levels of other model components, making it an attractive target for intervention. Overall, this model represents a step forward in the quantitative understanding of the dynamics of the heat-shock response.


Assuntos
Células HeLa/metabolismo , Resposta ao Choque Térmico/fisiologia , Modelos Biológicos , Simulação por Computador , DNA/metabolismo , Febre/metabolismo , Proteínas de Choque Térmico/metabolismo , Homeostase/fisiologia , Humanos , RNA Mensageiro/metabolismo , Temperatura , Fatores de Tempo
19.
Biochem Biophys Res Commun ; 467(4): 771-7, 2015 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-26482850

RESUMO

Considering the number of cytosolic proteins associated with many diseases, development of cytosol-penetrating molecules from outside of living cells is highly in demand. To gain access to the cytosol after cellular uptake, cell-penetrating molecules should be released from intermediate endosomes prior to the lysosomal degradation. However, it is very challenging to distinguish the pool of cytosolic-released molecules from those trapped in the endocytic vesicles. Here we describe a method to directly demonstrate the cytosolic localization and quantification of cytosolic amount of a cytosol-penetrating IgG antibody, TMab4, based on enhanced split GFP complementation system. We generated TMab4 genetically fused with one GFP fragment and separately established HeLa cells expressing the other GFP fragment in the cytosol such that the complemented GFP fluorescence is observed only when extracellular-treated TMab4 reaches the cytosol after cellular internalization. The high affinity interactions between streptavidin-binding peptide 2 and streptavidin was employed as respective fusion partners of GFP fragments to enhance the sensitivity of GFP complementation. With this method, cytosolic concentration of TMab4 was estimated to be about 170 nM after extracellular treatment of HeLa cells with 1 µM TMab4 for 6 h. We also found that after cellular internalization into living cells, nearly 1.3-4.3% of the internalized TMab4 molecules escaped into the cytosol from the endocytic vesicles. Our enhanced split GFP complementation assay provides a useful tool to directly quantify cytosolic amount of cytosol-penetrating agents and allows cell-based high-throughput screening for cytosol-penetrating agents with increased endosomal-escaping activity.


Assuntos
Anticorpos Monoclonais/metabolismo , Bioensaio/métodos , Citosol/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Imunoglobulina G/metabolismo , Sequência de Aminoácidos , Anticorpos Monoclonais/análise , Anticorpos Monoclonais/genética , Western Blotting , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Endossomos/metabolismo , Proteínas de Fluorescência Verde/genética , Células HEK293/metabolismo , Células HeLa/metabolismo , Humanos , Dados de Sequência Molecular , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Espectrometria de Fluorescência/métodos
20.
Biochem Biophys Res Commun ; 464(3): 922-8, 2015 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-26188516

RESUMO

Based on its specific interaction with cullin3 mediated by an N-terminal BTB/POZ homologous domain, KCTD5 has been proposed to function as substrate adapter for cullin3 based ubiquitin E3 ligases. In the present study we tried to validate this hypothesis through identification and characterization of additional KCTD5 interaction partners. For the replication protein MCM7, the zinc finger protein ZNF711 and FAM193B, a yet poorly characterized cytoplasmic protein, we could demonstrate specific interaction with KCTD5 both in yeast two-hybrid and co-precipitation studies in mammalian cells. Whereas trimeric complexes of cullin3 and KCTD5 with the respective KCTD5 binding partner were formed, KCTD5/cullin3 induced polyubiquitylation and/or proteasome-dependent degradation of these binding partners could not be demonstrated. On the contrary, KCTD5 or Cullin3 overexpression increased ZNF711 protein stability.


Assuntos
Proteínas Culina/metabolismo , Canais de Potássio/metabolismo , Divisão Celular , Citoplasma/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Células HeLa/metabolismo , Humanos , Componente 7 do Complexo de Manutenção de Minicromossomo/genética , Componente 7 do Complexo de Manutenção de Minicromossomo/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Canais de Potássio/genética , Mapeamento de Interação de Proteínas , Estabilidade Proteica , Transporte Proteico , Técnicas do Sistema de Duplo-Híbrido , Ubiquitinação
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa