Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
IEEE Trans Med Imaging ; 43(1): 459-472, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37647175

RESUMO

Self-supervised pretraining attempts to enhance model performance by obtaining effective features from unlabeled data, and has demonstrated its effectiveness in the field of histopathology images. Despite its success, few works concentrate on the extraction of nucleus-level information, which is essential for pathologic analysis. In this work, we propose a novel nucleus-aware self-supervised pretraining framework for histopathology images. The framework aims to capture the nuclear morphology and distribution information through unpaired image-to-image translation between histopathology images and pseudo mask images. The generation process is modulated by both conditional and stochastic style representations, ensuring the reality and diversity of the generated histopathology images for pretraining. Further, an instance segmentation guided strategy is employed to capture instance-level information. The experiments on 7 datasets show that the proposed pretraining method outperforms supervised ones on Kather classification, multiple instance learning, and 5 dense-prediction tasks with the transfer learning protocol, and yields superior results than other self-supervised approaches on 8 semi-supervised tasks. Our project is publicly available at https://github.com/zhiyuns/UNITPathSSL.

2.
Front Immunol ; 13: 828560, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35464416

RESUMO

Background: Programmed death-ligand 1 (PD-L1) assessment of lung cancer in immunohistochemical assays was only approved diagnostic biomarker for immunotherapy. But the tumor proportion score (TPS) of PD-L1 was challenging owing to invasive sampling and intertumoral heterogeneity. There was a strong demand for the development of an artificial intelligence (AI) system to measure PD-L1 expression signature (ES) non-invasively. Methods: We developed an AI system using deep learning (DL), radiomics and combination models based on computed tomography (CT) images of 1,135 non-small cell lung cancer (NSCLC) patients with PD-L1 status. The deep learning feature was obtained through a 3D ResNet as the feature map extractor and the specialized classifier was constructed for the prediction and evaluation tasks. Then, a Cox proportional-hazards model combined with clinical factors and PD-L1 ES was utilized to evaluate prognosis in survival cohort. Results: The combination model achieved a robust high-performance with area under the receiver operating characteristic curves (AUCs) of 0.950 (95% CI, 0.938-0.960), 0.934 (95% CI, 0.906-0.964), and 0.946 (95% CI, 0.933-0.958), for predicting PD-L1ES <1%, 1-49%, and ≥50% in validation cohort, respectively. Additionally, when combination model was trained on multi-source features the performance of overall survival evaluation (C-index: 0.89) could be superior compared to these of the clinical model alone (C-index: 0.86). Conclusion: A non-invasive measurement using deep learning was proposed to access PD-L1 expression and survival outcomes of NSCLC. This study also indicated that deep learning model combined with clinical characteristics improved prediction capabilities, which would assist physicians in making rapid decision on clinical treatment options.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Aprendizado Profundo , Neoplasias Pulmonares , Algoritmos , Inteligência Artificial , Antígeno B7-H1/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Humanos , Neoplasias Pulmonares/patologia
3.
Proc Natl Acad Sci U S A ; 116(19): 9568-9577, 2019 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-31019079

RESUMO

Lentiviruses, including HIV-1, possess the ability to enter the nucleus through nuclear pore complexes and can infect interphase cells, including those actively replicating chromosomal DNA. Viral accessory proteins hijack host cell E3 enzymes to antagonize intrinsic defenses, and thereby provide a more permissive environment for virus replication. The HIV-1 Vpr accessory protein reprograms CRL4DCAF1 E3 to antagonize select postreplication DNA repair enzymes and activates the DNA damage checkpoint in the G2 cell cycle phase. However, little is known about the roles played by these Vpr targets in HIV-1 replication. Here, using a sensitive pairwise replication competition assay, we show that Vpr endows HIV-1 with a strong replication advantage in activated primary CD4+ T cells and established T cell lines. This effect is disabled by a Vpr mutation that abolishes binding to CRL4DCAF1 E3, thereby disrupting Vpr antagonism of helicase-like transcription factor (HLTF) DNA helicase and other DNA repair pathway targets, and by another mutation that prevents induction of the G2 DNA damage checkpoint. Consistent with these findings, we also show that HLTF restricts HIV-1 replication, and that this restriction is antagonized by HIV-1 Vpr. Furthermore, our data imply that HIV-1 Vpr uses additional, yet to be identified mechanisms to facilitate HIV-1 replication in T cells. Overall, we demonstrate that multiple aspects of the cellular DNA repair machinery restrict HIV-1 replication in dividing T cells, the primary target of HIV-1 infection, and describe newly developed approaches to dissect key components.


Assuntos
Linfócitos T CD4-Positivos , Proteínas de Ligação a DNA/metabolismo , Infecções por HIV , HIV-1/fisiologia , Fatores de Transcrição/metabolismo , Replicação Viral/fisiologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD4-Positivos/virologia , Proteínas de Ligação a DNA/genética , Pontos de Checagem da Fase G2 do Ciclo Celular , Infecções por HIV/genética , Infecções por HIV/metabolismo , Infecções por HIV/patologia , Humanos , Fatores de Transcrição/genética , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/genética
4.
mBio ; 9(5)2018 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-30352932

RESUMO

Viral accessory proteins hijack host cell E3 ubiquitin ligases to antagonize innate/intrinsic defenses and thereby provide a more permissive environment for virus replication. Human immunodeficiency virus type 1 (HIV-1) accessory protein Vpr reprograms CRL4DCAF1 E3 to antagonize select postreplication DNA repair enzymes, but the significance and role of these Vpr interactions are poorly understood. To gain additional insights, we performed a focused screen for substrates of CRL4DCAF1 E3 reprogrammed by HIV-1 Vpr among known postreplication DNA repair proteins and identified exonuclease 1 (Exo1) as a novel direct HIV-1 Vpr target. We show that HIV-1 Vpr recruits Exo1 to the CRL4DCAF1 E3 complex for ubiquitination and subsequent proteasome-dependent degradation and that Exo1 levels are depleted in HIV-1-infected cells in a Vpr-dependent manner. We also show that Exo1 inhibits HIV-1 replication in T cells. Notably, the antagonism of Exo1 is a conserved function of main group HIV-1 and its ancestor Vpr proteins in the simian immunodeficiency virus from chimpanzee (SIVcpz) lineage, further underscoring the relevance of our findings. Overall, our studies (i) reveal that HIV-1 Vpr extensively remodels the cellular postreplication DNA repair machinery by impinging on multiple repair pathways, (ii) support a model in which Vpr promotes HIV-1 replication by antagonizing select DNA repair enzymes, and (iii) highlight the importance of a new class of restrictions placed on HIV-1 replication in T cells by the cellular DNA repair machinery.IMPORTANCE HIV-1 polymerase reverse transcribes the viral RNA genome into imperfectly double-stranded proviral DNA, containing gaps and flaps, for integration into the host cell chromosome. HIV-1 reverse transcripts share characteristics with cellular DNA replication intermediates and are thought to be converted into fully double-stranded DNA by cellular postreplication DNA repair enzymes. Therefore, the finding that the HIV-1 accessory protein Vpr antagonizes select postreplication DNA repair enzymes that can process HIV-1 reverse transcripts has been surprising. Here, we show that one such Vpr-antagonized enzyme, exonuclease 1, inhibits HIV-1 replication in T cells. We identify exonuclease 1 as a member of a new class of HIV-1 restriction factors in T cells and propose that certain modes of DNA "repair" inhibit HIV-1 infection.


Assuntos
Enzimas Reparadoras do DNA/metabolismo , Exodesoxirribonucleases/metabolismo , Interações Hospedeiro-Patógeno , Ubiquitina-Proteína Ligases/metabolismo , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo , Reparo do DNA , Enzimas Reparadoras do DNA/antagonistas & inibidores , Enzimas Reparadoras do DNA/genética , Proteínas de Ligação a DNA/metabolismo , Exodesoxirribonucleases/antagonistas & inibidores , Exodesoxirribonucleases/genética , Infecções por HIV/enzimologia , Infecções por HIV/genética , HIV-1/genética , HIV-1/fisiologia , Humanos , Linfócitos T/virologia , Ubiquitina-Proteína Ligases/genética , Ubiquitinação , Replicação Viral , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/genética
5.
Biomed Res Int ; 2015: 739586, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26504826

RESUMO

BACKGROUND: To investigate whether one or more SAMHD1 gene mutations are associated with cerebrovascular disease in the general population using a Chinese stroke cohort. METHODS: Patients with a Chinese Han background (N = 300) diagnosed with either cerebral large-artery atherosclerosis (LAA, n = 100), cerebral small vessel disease (SVD, n = 100), or other stroke-free neurological disorders (control, n = 100) were recruited. Genomic DNA from the whole blood of each patient was isolated, and direct sequencing of the SAMHD1 gene was performed. Both wild type and mutant SAMHD1 proteins identified from the patients were expressed in E. coli and purified; then their dNTPase activities and ability to form stable tetramers were analysed in vitro. RESULTS: Three heterozygous mutations, including two missense mutations c.64C>T (P22S) and c.841G>A (p.E281K) and one splice site mutation c.696+2T>A, were identified in the LAA group with a prevalence of 3%. No mutations were found in the patients with SVD or the controls (p = 0.05). The mutant SAMHD1 proteins were functionally impaired in terms of their catalytic activity as a dNTPase and ability to assemble stable tetramers. CONCLUSIONS: Heterozygous SAMHD1 gene mutations might cause genetic predispositions that interact with other risk factors, resulting in increased vulnerability to stroke.


Assuntos
Aterosclerose/genética , Predisposição Genética para Doença/genética , Proteínas Monoméricas de Ligação ao GTP/genética , Acidente Vascular Cerebral/genética , Idoso , Povo Asiático/genética , Aterosclerose/epidemiologia , Artérias Cerebrais/diagnóstico por imagem , China/epidemiologia , Estudos de Coortes , Análise Mutacional de DNA , Feminino , Predisposição Genética para Doença/epidemiologia , Humanos , Masculino , Pessoa de Meia-Idade , Mutação/genética , Radiografia , Proteína 1 com Domínio SAM e Domínio HD , Acidente Vascular Cerebral/epidemiologia
6.
J Biol Chem ; 290(21): 13279-92, 2015 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-25847232

RESUMO

SAMHD1 is a nuclear deoxyribonucleoside triphosphate triphosphohydrolase that contributes to the control of cellular deoxyribonucleoside triphosphate (dNTP) pool sizes through dNTP hydrolysis and modulates the innate immune response to viruses. CyclinA2-CDK1/2 phosphorylates SAMHD1 at Thr-592, but how this modification controls SAMHD1 functions in proliferating cells is not known. Here, we show that SAMHD1 levels remain relatively unchanged during the cell division cycle in primary human T lymphocytes and in monocytic cell lines. Inactivation of the bipartite cyclinA2-CDK-binding site in the SAMHD1 C terminus described herein abolished SAMHD1 phosphorylation on Thr-592 during S and G2 phases thus interfering with DNA replication and progression of cells through S phase. The effects exerted by Thr-592 phosphorylation-defective SAMHD1 mutants were associated with activation of DNA damage checkpoint and depletion of dNTP concentrations to levels lower than those seen upon expression of wild type SAMHD1 protein. These disruptive effects were relieved by either mutation of the catalytic residues of the SAMHD1 phosphohydrolase domain or by a Thr-592 phosphomimetic mutation, thus linking the Thr-592 phosphorylation state to the control of SAMHD1 dNTPase activity. Our findings support a model in which phosphorylation of Thr-592 by cyclinA2-CDK down-modulates, but does not inactivate, SAMHD1 dNTPase in S phase, thereby fine-tuning SAMHD1 control of dNTP levels during DNA replication.


Assuntos
Ciclo Celular/fisiologia , Ciclina A2/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Monócitos/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Linfócitos T/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Western Blotting , Proteína Quinase CDC2 , Células Cultivadas , Ciclina A2/genética , Quinase 2 Dependente de Ciclina/genética , Quinases Ciclina-Dependentes/genética , Células HEK293 , Humanos , Hidrólise , Técnicas Imunoenzimáticas , Imunoprecipitação , Dados de Sequência Molecular , Monócitos/citologia , Proteínas Monoméricas de Ligação ao GTP/genética , Mutação/genética , Fosforilação , Estrutura Terciária de Proteína , Proteína 1 com Domínio SAM e Domínio HD , Homologia de Sequência de Aminoácidos , Linfócitos T/citologia
7.
Viruses ; 6(8): 3228-49, 2014 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-25140737

RESUMO

The cellular bromodomain protein Brd4 functions in multiple processes of the papillomavirus life cycle, including viral replication, genome maintenance, and gene transcription through its interaction with the viral protein, E2. However, the mechanisms by which E2 and Brd4 activate viral transcription are still not completely understood. In this study, we show that recruitment of positive transcription elongation factor b (P-TEFb), a functional interaction partner of Brd4 in transcription activation, is important for E2's transcription activation activity. Furthermore, chromatin immunoprecipitation (ChIP) analyses demonstrate that P-TEFb is recruited to the actual papillomavirus episomes. We also show that E2's interaction with cellular chromatin through Brd4 correlates with its papillomavirus transcription activation function since JQ1(+), a bromodomain inhibitor that efficiently dissociates E2-Brd4 complexes from chromatin, potently reduces papillomavirus transcription. Our study identifies a specific function of Brd4 in papillomavirus gene transcription and highlights the potential use of bromodomain inhibitors as a method to disrupt the human papillomavirus (HPV) life cycle.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Interações Hospedeiro-Patógeno , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas Virais/metabolismo , Proteínas Oncogênicas Virais/fisiologia , Fatores de Transcrição/metabolismo , Ativação Transcricional , Proteínas de Ciclo Celular , Linhagem Celular , Imunoprecipitação da Cromatina , DNA Viral/metabolismo , Humanos , Fator B de Elongação Transcricional Positiva/metabolismo
8.
Nat Struct Mol Biol ; 20(11): 1304-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24141705

RESUMO

SAMHD1, a dNTP triphosphohydrolase (dNTPase), has a key role in human innate immunity. It inhibits infection of blood cells by retroviruses, including HIV, and prevents the development of the autoinflammatory Aicardi-Goutières syndrome (AGS). The inactive apo-SAMHD1 interconverts between monomers and dimers, and in the presence of dGTP the protein assembles into catalytically active tetramers. Here, we present the crystal structure of the human tetrameric SAMHD1-dGTP complex. The structure reveals an elegant allosteric mechanism of activation through dGTP-induced tetramerization of two inactive dimers. Binding of dGTP to four allosteric sites promotes tetramerization and induces a conformational change in the substrate-binding pocket to yield the catalytically active enzyme. Structure-based biochemical and cell-based biological assays confirmed the proposed mechanism. The SAMHD1 tetramer structure provides the basis for a mechanistic understanding of its function in HIV restriction and the pathogenesis of AGS.


Assuntos
Regulação Alostérica , Nucleotídeos de Desoxiguanina/química , Nucleotídeos de Desoxiguanina/metabolismo , Proteínas Monoméricas de Ligação ao GTP/química , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Multimerização Proteica , Sítios de Ligação , Cristalografia por Raios X , Humanos , Modelos Moleculares , Conformação Proteica , Proteína 1 com Domínio SAM e Domínio HD
9.
J Biol Chem ; 288(15): 10406-17, 2013 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-23426366

RESUMO

SAMHD1 is a dGTP-activated dNTPase that has been implicated as a modulator of the innate immune response. In monocytes and their differentiated derivatives, as well as in quiescent cells, SAMHD1 strongly inhibits HIV-1 infection and, to a lesser extent, HIV-2 and simian immunodeficiency virus (SIV) because of their virion-associated virulence factor Vpx, which directs SAMHD1 for proteasomal degradation. Here, we used a combination of biochemical and virologic approaches to gain insights into the functional organization of human SAMHD1. We found that the catalytically active recombinant dNTPase is a dGTP-induced tetramer. Chemical cross-linking studies revealed SAMHD1 tetramers in human monocytic cells, in which it strongly restricts HIV-1 infection. The propensity of SAMHD1 to maintain the tetrameric state in vitro is regulated by its C terminus, located outside of the catalytic domain. Accordingly, we show that the C terminus is required for the full ability of SAMHD1 to deplete dNTP pools and to inhibit HIV-1 infection in U937 monocytes. Interestingly, the human SAMHD1 C terminus contains a docking site for HIV-2/SIVmac Vpx and is known to have evolved under positive selection. This evidence indicates that Vpx targets a functionally important element in SAMHD1. Together, our findings imply that SAMHD1 tetramers are the biologically active form of this dNTPase and provide new insights into the functional organization of SAMHD1.


Assuntos
Infecções por HIV/enzimologia , HIV-1/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Multimerização Proteica , Infecções por HIV/genética , HIV-1/genética , HIV-2/genética , HIV-2/metabolismo , Humanos , Proteínas Monoméricas de Ligação ao GTP/genética , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Proteólise , Proteína 1 com Domínio SAM e Domínio HD , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/metabolismo , Células U937
10.
J Biol Chem ; 287(15): 12550-8, 2012 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-22362772

RESUMO

The sterile alpha motif and HD domain-containing protein-1 (SAMHD1) inhibits infection of myeloid cells by human and related primate immunodeficiency viruses (HIV and SIV). This potent inhibition is counteracted by the Vpx accessory virulence factor of HIV-2/SIVsm viruses, which targets SAMHD1 for proteasome-dependent degradation, by reprogramming cellular CRL4(DCAF1) E3 ubiquitin ligase. However, the precise mechanism of Vpx-dependent recruitment of human SAMHD1 onto the ligase, and the molecular interfaces on the respective molecules have not been defined. Here, we show that human SAMHD1 is recruited to the CRL4(DCAF1-Vpx) E3 ubiquitin ligase complex by interacting with the DCAF1 substrate receptor subunit in a Vpx-dependent manner. No stable association is detectable with DCAF1 alone. The SAMHD1 determinant for the interaction is a short peptide located distal to the SAMHD1 catalytic domain and requires the presence of Vpx for stable engagement. This peptide is sufficient to confer Vpx-dependent recruitment to CRL4(DCAF1) and ubiquitination when fused to heterologous proteins. The precise amino acid sequence of the peptide diverges among SAMHD1 proteins from different vertebrate species, explaining selective down-regulation of human SAMHD1 levels by Vpx. Critical amino acid residues of SAMHD1 and Vpx involved in the DCAF1-Vpx-SAMDH1 interaction were identified by mutagenesis. Our findings show that the N terminus of Vpx, bound to DCAF1, recruits SAMHD1 via its C terminus to CRL4, in a species-specific manner for proteasomal degradation.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Proteínas Nucleares/metabolismo , Vírus da Imunodeficiência Símia/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Sequência Conservada , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Dados de Sequência Molecular , Proteínas Monoméricas de Ligação ao GTP/genética , Mutagênese Sítio-Dirigida , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas Serina-Treonina Quinases , Proteínas Recombinantes de Fusão/metabolismo , Proteína 1 com Domínio SAM e Domínio HD
11.
J Biol Chem ; 286(31): 27663-75, 2011 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-21652721

RESUMO

NUT midline carcinoma (NMC) belongs to a class of highly lethal and poorly differentiated epithelial cancers arising mainly in human midline organs. NMC is caused by the chromosome translocation-mediated fusion of the NUT (nuclear protein in testis) gene on chromosome 15 to a few other genes, most frequently the BRD4 gene on chromosome 19. The mechanism by which the BRD4-NUT fusion product blocks NMC cellular differentiation and contributes to oncogenesis remains elusive. In this study, we show that BRD4-NUT and BRD4 colocalize in discrete nuclear foci that are hyperacetylated but transcriptionally inactive. BRD4-NUT recruits histone acetyltransferases to induce histone hyperacetylation in these chromatin foci, which provide docking sites for accumulation of additional BRD4 and associated P-TEFB (positive transcription elongation factor b) complexes in the transcriptionally inactive BRD4-NUT foci. These molecular events lead to repression of a BRD4·P-TEFB downstream target gene c-fos, a component of activator protein 1 (AP-1), that directly regulates epithelial differentiation. Knockdown of BRD4-NUT in NMC cells disperses the transcriptionally inactive chromatin foci and releases the transcriptional activators to stimulate c-fos expression, leading to restoration of cellular differentiation. Our study provides a novel mechanism by which the BRD4-NUT oncogene perturbs BRD4 functions to block cellular differentiation and to contribute to the oncogenic progression in the highly aggressive NMC.


Assuntos
Carcinoma/patologia , Diferenciação Celular/fisiologia , Proteínas Nucleares/fisiologia , Proteínas Oncogênicas/fisiologia , Fatores de Transcrição/fisiologia , Acetilação , Carcinoma/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Regulação para Baixo , Genes fos , Humanos , Imunoprecipitação , Proteínas de Neoplasias , Proteínas Nucleares/genética , Proteínas Oncogênicas/genética , Plasmídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
J Microbiol ; 48(2): 199-205, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20437152

RESUMO

The inhibitor of apoptosis proteins (IAP) plays an important role in cell apoptosis. We cloned two novel IAP family members, Ap-iap1 and Ap-iap2, from Antheraea pernyi nucleopolyhedrovirus (ApNPV) genome. Ap-IAP1 contains two baculoviral IAP repeat (BIR) domains followed by a RING domain, but Ap-IAP2 has only one BIR domain and RING. The result of transient expression in Spodoptera frugiperda (Sf21) showed that Ap-iap1 blocked cell apoptosis induced by actinomycin D treatment and also rescued the p35 deficient Autographa californica nucleopolyhedrovirus (AcNPV) to replicate in Sf9 cells, while Ap-iap2 does not have this function. Several Ap-IAP1 truncations were constructed to test the activity of BIRs or RING motif to inhibit cell apoptosis. The results indicated that BIRs or RING of Ap-IAP1 had equally function to inhibit cell apoptosis. Therefore deletion of above both of the above domains could not block apoptosis induced by actinomycin D or rescue the replication of AcMNPV Delta p35. We also screened two phage-display peptides that might interact with Ap-IAP1.


Assuntos
Proteínas Inibidoras de Apoptose/fisiologia , Mariposas/virologia , Nucleopoliedrovírus/patogenicidade , Proteínas Virais/fisiologia , Fatores de Virulência/fisiologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Linhagem Celular , Clonagem Molecular , Proteínas Inibidoras de Apoptose/genética , Dados de Sequência Molecular , Alinhamento de Sequência , Deleção de Sequência , Spodoptera/virologia , Proteínas Virais/genética , Fatores de Virulência/genética
13.
J Virol ; 84(1): 76-87, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19846528

RESUMO

The cellular bromodomain protein Brd4 is a major interacting partner of the papillomavirus (PV) E2 protein. Interaction of E2 with Brd4 contributes to viral episome maintenance. The E2-Brd4 interaction also plays an important role in repressing viral oncogene expression from the integrated viral genome in human PV (HPV)-positive cancer cells. However, the underlying mechanism is not clearly understood. In host cells, Brd4 recruits positive transcription elongation factor b (P-TEFb) to stimulate RNA polymerase II phosphorylation during cellular and viral gene expression. P-TEFb associates with the C terminus of Brd4, which largely overlaps with the E2 binding site on Brd4. In this study, we demonstrate that E2 binding to Brd4 inhibits the interaction of endogenous Brd4 and P-TEFb. P-TEFb is essential for viral oncogene E6/E7 transcription in both HeLa and CaSki cells that contain integrated HPV genomes. E2 binding to Brd4 abrogates the recruitment of P-TEFb to the integrated viral chromatin template, leading to inactivation of P-TEFb and repression of the viral oncogene E6/E7. Furthermore, dissociation of the Brd4-P-TEFb complex from the integrated viral chromatin template using a Brd4 bromodomain dominant-negative inhibitor also hampers HPV E6/E7 oncogene expression. Our data support that Brd4 recruitment of P-TEFb to the viral chromatin template is essential for viral oncogene expression. Abrogation of the interaction between P-TEFb and Brd4 thus provides a mechanism for E2-mediated repression of the viral oncogenes from the integrated viral genomes in cancer cells.


Assuntos
Alphapapillomavirus/genética , Proteínas de Ligação a DNA/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas Virais/genética , Papillomaviridae/fisiologia , Fator B de Elongação Transcricional Positiva/antagonistas & inibidores , Fatores de Transcrição/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular , Linhagem Celular Tumoral , Genes Virais , Humanos , Proteínas Oncogênicas Virais/antagonistas & inibidores , Proteínas Oncogênicas Virais/fisiologia , Papillomaviridae/química , Proteínas E7 de Papillomavirus , Fator B de Elongação Transcricional Positiva/genética , Fator B de Elongação Transcricional Positiva/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Transcrição Gênica
14.
J Thromb Thrombolysis ; 28(4): 444-8, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19377880

RESUMO

Subtilisin QK, a new fibrinolytic enzyme, could cleave directly cross-linked fibrin in vitro. To verify the thrombolytic function of Subtilisin QK in vivo, the thrombolytic effect of purified Subtilisin QK on tail-thrombus of mouse was investigated. After injected with carrageenan, the tail-thrombus of Subtilisin QK treated group were shorter than the physiological saline treated group. Moreover, the tail-thrombus decreased correlate with Subtilisin QK in a dose-dependent manner. Thrombus nearly disappeared while the mice were treated with 12000 IU Subtilisin QK. The result indicated that Subtilisin QK significantly inhibited thrombus formation in mouse tail. This study made more foundation for further development of Subtilisin QK as a novel bifunctional thrombolytic agent.


Assuntos
Carragenina/toxicidade , Modelos Animais de Doenças , Fibrinolíticos/uso terapêutico , Subtilisinas/uso terapêutico , Trombose/tratamento farmacológico , Trombose/enzimologia , Animais , Relação Dose-Resposta a Droga , Humanos , Masculino , Camundongos , Trombose/induzido quimicamente
15.
J Microbiol Biotechnol ; 17(4): 594-9, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18051269

RESUMO

It is well known that the Escherichia coli inner membrane-bound protease DegS is a periplasmic stress sensor for unfolded outer membrane proteins (OMPs). Previous studies have also shown that the outer membrane protease OmpT activates plasminogen in vitro and this may be exploited by bacteria in the course of pathogenesis. However, there has been no research on the plasminogen activation ability of the important periplasmic protein DegS. Accordingly, in this study, the whole-length and truncated degS genes were separately overexpressed in Escherichia coli, the recombinant proteins purified by affinity chromatography, and their plasminogen activator role tested in vitro. The results suggested that the whole-length DegS was able to activate plasminogen on a plasma plate. The truncated form of DegS (residues 80-345), designated delta DegS, also acted as a plasminogen activator, as confirmed by different assays. The serine protease property of delta DegS was verified based on the complete inhibition of its enzyme activity by PMSF (phenylmethanesulfonyl fluoride). Therefore, the present results indicate that DegS is a plasminogen activator in vitro.


Assuntos
Proteínas de Escherichia coli/farmacologia , Fragmentos de Peptídeos/farmacologia , Ativadores de Plasminogênio/farmacologia , Proteínas de Escherichia coli/química , Humanos
16.
J Biochem Mol Biol ; 38(5): 577-83, 2005 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-16202238

RESUMO

Subtilisin QK, which is newly identified as a fibrinolytic enzyme from Bacillus subtilis QK02, has the ability of preventing nitrotyrosine formation in bovine serum albumin induced by nitrite, hydrogen peroxide and hemoglobin in vitro verified by ELISA, Western-blot and spectrophotometer assay. Subtilisin QK also attenuates the fluorescence emission spectra of bovine serum albumin in the course of oxidation caused by nitrite, hydrogen peroxide and hemoglobin. Furthermore, subtilisin QK could suppress the transformation of oxy-hemoglobin to met-hemoglobin caused by sodium nitrite, but not the heat-treated subtilisn QK. Compared with some other fibrinolytic enzymes and inactivated subtilisin QK treated by phenylmethylsulfonylfluoride, the ability of inhibiting met-hemoglobin formation of subtilisin QK reveals that the anti-oxidative ability of subtilisin QK is not concerned with its fibrinolytic function. Additionally, nitrotyrosine formation in proteins from brain, heart, liver, kidney, and muscle of mice that is intramuscular injected the mixture of nitrite, hydrogen peroxide and hemoglobin is attenuated by subtilisin QK. Subtilisin QK can also protect Human umbilical vein endothelial cell (ECV-304) from the damage caused by nitrite and hydrogen peroxide.


Assuntos
Peróxido de Hidrogênio/metabolismo , Nitratos/metabolismo , Nitritos/metabolismo , Oxidantes/metabolismo , Soroalbumina Bovina/metabolismo , Subtilisinas/metabolismo , Animais , Antioxidantes/metabolismo , Bacillus subtilis/enzimologia , Bovinos , Linhagem Celular , Hemoglobinas/metabolismo , Humanos , Masculino , Camundongos , Oxirredução , Ratos , Tirosina/química
17.
Biochem Biophys Res Commun ; 329(2): 437-44, 2005 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-15737606

RESUMO

To develop early diagnostic reagents, effective vaccines, and even drugs against SARS-associated coronavirus (SARS-CoV), the human single fold single-chain antibody fragments, (scFv) libraries I+J (Tomlinson I+J) were used to identify novel scFvs, which can specifically bind to SARS-CoV. Interestingly, two scFvs (B5 and B9) exhibited higher binding specificity to SARS-CoV with the OD(450) value 0.608 and 0.545, respectively, and their coding sequences shared the identical sequence composed of V(H) gene (351bp) and V(L) gene (327bp), so the two scFvs were uniformly named as SA59B and chosen for further analysis. SA59B scFv was expressed in soluble form in Escherichia coli HB2151 and purified by immobilized metal affinity chromatography. The soluble 30kDa SA59B scFv-antibody was verified in SDS-PAGE and Western-blot. The purified SA59B scFv-antibody was labeled with HRP by the glutaraldehyde method, and the concentration of HRP and SA59B scFv-antibody in the SA59B-HRP solution reached 2.4 and 2.28mg/ml, respectively. Then, the binding ability of SA59B-HRP to SARS-CoV was evaluated by ELISA with S/N of 11.6, indicating higher binding specificity between them. Finally, both the SA59B sequence specificity and its application for diagnosis, prophylaxis or therapy of SARS were discussed.


Assuntos
Ensaio de Imunoadsorção Enzimática/métodos , Fragmentos de Imunoglobulinas/química , Fragmentos de Imunoglobulinas/imunologia , Biblioteca de Peptídeos , Análise de Sequência/métodos , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/imunologia , Sequência de Aminoácidos , Fragmentos de Imunoglobulinas/análise , Dados de Sequência Molecular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...