Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Virol ; 97(10): e0063723, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37750723

RESUMO

IMPORTANCE: Kaposi's sarcoma-associated herpesvirus (KSHV) is a human herpesvirus associated with several human cancers, typically in patients with compromised immune systems. Herpesviruses establish lifelong infections in hosts in part due to the two phases of infection: the dormant and active phases. Effective antiviral treatments to prevent the production of new viruses are needed to treat KSHV. A detailed microscopy-based investigation of the molecular interactions between viral protein and viral DNA revealed how protein-protein interactions play a role in DNA-binding specificity. This analysis will lead to a more in-depth understanding of KSHV DNA replication and serve as the basis for anti-viral therapies that disrupt and prevent the protein-DNA interactions, thereby decreasing spread to new hosts.


Assuntos
DNA Viral , Herpesvirus Humano 8 , Microscopia Eletrônica , Multimerização Proteica , Transativadores , Humanos , Sítios de Ligação , DNA Viral/química , DNA Viral/metabolismo , DNA Viral/ultraestrutura , Herpesvirus Humano 8/química , Herpesvirus Humano 8/metabolismo , Herpesvirus Humano 8/ultraestrutura , Ligação Proteica , Mapas de Interação de Proteínas , Especificidade por Substrato , Transativadores/química , Transativadores/metabolismo , Transativadores/ultraestrutura , Replicação Viral/genética , Sarcoma de Kaposi/virologia
2.
bioRxiv ; 2023 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-37205529

RESUMO

Molecular interactions between viral DNA and viral-encoded protein are a prerequisite for successful herpesvirus replication and production of new infectious virions. Here, we examined how the essential Kaposi's sarcoma-associated herpesvirus (KSHV) protein, RTA, binds to viral DNA using transmission electron microscopy (TEM). Previous studies using gel-based approaches to characterize RTA binding are important for studying the predominant form(s) of RTA within a population and identifying the DNA sequences that RTA binds with high affinity. However, using TEM we were able to examine individual protein-DNA complexes and capture the various oligomeric states of RTA when bound to DNA. Hundreds of images of individual DNA and protein molecules were collected and then quantified to map the DNA binding positions of RTA bound to the two KSHV lytic origins of replication encoded within the KSHV genome. The relative size of RTA or RTA bound to DNA were then compared to protein standards to determine whether RTA complexed with DNA was monomeric, dimeric, or formed larger oligomeric structures. We successfully analyzed a highly heterogenous dataset and identified new binding sites for RTA. This provides direct evidence that RTA forms dimers and high order multimers when bound to KSHV origin of replication DNA sequences. This work expands our understanding of RTA binding, and demonstrates the importance of employing methodologies that can characterize highly heterogenic populations of proteins. Importance: Kaposi's sarcoma-associated herpesvirus (KSHV) is a human herpesvirus associated with several human cancers, typically in patients with compromised immune systems. Herpesviruses establish lifelong infections in hosts in part due to the two phases of infection: the dormant and active phases. Effective antiviral treatments to prevent the production of new viruses are needed to treat KSHV. A detailed microscopy-based investigation of the molecular interactions between viral protein and viral DNA revealed how protein-protein interactions play a role in DNA binding specificity. This analysis will lead to a more in depth understanding of KSHV DNA replication and serve as the basis for anti-viral therapies that disrupt and prevent the protein-DNA interactions, thereby decreasing spread to new hosts.

3.
ACS Bio Med Chem Au ; 2(2): 140-149, 2022 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-35480227

RESUMO

Extracellular vesicles (EVs), or exosomes, play a pivotal role in tumor growth and metastasis, such as in the case of Kaposi Sarcoma. By loading tumor-derived EVs with chemotherapeutic drugs, we noted that their pro-tumor/pro-angiogenic phenotype was converted into an anti-tumor phenotype in vivo. Drug concentration in EVs was significantly higher than in clinically approved liposome formulation, as retention was facilitated by the presence of miRNAs inside the natural EVs. This demonstrates a new mechanism by which to increase the payload capacity of nanoparticles. By exploiting the targeting preferences of tumor-derived EVs, chemotherapeutics can be directed to specifically poison the cells and the microenvironment that enables metastasis.

4.
J Extracell Vesicles ; 11(3): e12191, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35234354

RESUMO

Extracellular vesicles (EVs) are secreted from all cell types and are intimately involved in tissue homeostasis. They are being explored as vaccine and gene therapy platforms, as well as potential biomarkers. As their size is below the diffraction limit of light microscopy, direct visualizations have been daunting and single-particle studies under physiological conditions have been hampered. Here, direct stochastic optical reconstruction microscopy (dSTORM) was employed to visualize EVs in three-dimensions and to localize molecule clusters such as the tetraspanins CD81 and CD9 on the surface of individual EVs. These studies demonstrate the existence of membrane microdomains on EVs. These were confirmed by Cryo-EM. Individual particle visualization provided insights into the heterogeneity, structure, and complexity of EVs not previously appreciated.


Assuntos
Vesículas Extracelulares , Transporte Biológico , Biomarcadores/análise , Vesículas Extracelulares/química , Microscopia , Tetraspaninas/análise
5.
Data Brief ; 26: 104464, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31667234

RESUMO

This data article is related to the research article entitled "Silver nanoparticles alter epithelial basement membrane integrity, cell adhesion molecule expression and TGF-beta secretion", available in the journal Nanomedicine: Nanotechnology, Biology, and Medicine [1]. This Data in Brief consists of data that describe changes in the expression of basement membrane (BM)-associated genes and proteins in three non-transformed epithelial cell lines following acute (6 h) and chronic (24 h plus 7-day chase) exposure to silver nanoparticles (AgNPs). Human BEAS2B (lung), MCF10AI (breast), and CCD-18Co (colon) cultured epithelia were analyzed for protein expression by LC-MS/MS and for gene expression by pathway-focused QRT-PCR arrays of 168 focal adhesion, integrin, and extracellular matrix (ECM) genes known to be localized to the plasma membrane, the BM/ECM, or secreted into the extracellular space. Ingenuity pathway analysis (IPA) of combined gene and protein expression datasets was then used to predict canonical pathways affected by AgNP exposure.

6.
Structure ; 27(10): 1527-1536.e3, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31422909

RESUMO

Vinculin and its splice isoform metavinculin play key roles in regulating cellular morphology, motility, and force transduction. Vinculin is distinct from metavinculin in its ability to bundle filamentous actin (F-actin). To elucidate the molecular basis for these differences, we employed computational and experimental approaches. Results from these analyses suggest that the C terminus of both vinculin and metavinculin form stable interactions with the F-actin surface. However, the metavinculin tail (MVt) domain contains a 68 amino acid insert, with helix 1 (H1) sequestered into a globular subdomain, which protrudes from the F-actin surface and prevents actin bundling by sterically occluding actin filaments. Consistent with our model, deletion and selective point mutations within the MVt H1 disrupt this protruding structure, and facilitate actin bundling similar to vinculin tail (Vt) domain.


Assuntos
Vinculina/química , Vinculina/metabolismo , Actinas , Processamento Alternativo , Animais , Sítios de Ligação , Microscopia Crioeletrônica , Modelos Moleculares , Mutação , Ligação Proteica , Domínios Proteicos , Estrutura Secundária de Proteína , Vinculina/genética
7.
Nanomedicine ; 21: 102070, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31351238

RESUMO

Silver nanoparticles (AgNPs) are widely used in consumer and pharmaceutical products due to their antipathogenic properties. However, safety concerns have been raised due to their bioactive properties. While reports have demonstrated AgNPs can embed within the extracellular matrix, their effects on basement membrane (BM) production, integrin engagement, and tissue-integrity are not well-defined. This study analyzed the effects of AgNPs on BM production, composition and integrin/focal adhesion interactions in representative lung, esophageal, breast and colorectal epithelia models. A multidisciplinary approach including focused proteomics, QPCR arrays, pathway analyses, and immune-based, structural and functional assays was used to identify molecular and physiological changes in cell adhesions and the BM induced by acute and chronic AgNP exposure. Dysregulated targets included CD44 and transforming growth factor-beta, two proteins frequently altered during pathogenesis. Results indicate AgNP exposure interferes with BM and cell adhesion dynamics, and provide insight into the mechanisms of AgNP-induced disruption of epithelial physiology.


Assuntos
Membrana Basal/metabolismo , Moléculas de Adesão Celular/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Nanopartículas Metálicas/química , Prata , Fator de Crescimento Transformador beta1/biossíntese , Linhagem Celular Tumoral , Humanos , Prata/química , Prata/farmacologia
8.
J Mol Biol ; 431(8): 1604-1618, 2019 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-30844403

RESUMO

Debilitating heart conditions, notably dilated and hypertrophic cardiomyopathies (CMs), are associated with point mutations in metavinculin, a larger isoform of the essential cytoskeletal protein vinculin. Metavinculin is co-expressed with vinculin at sub-stoichiometric ratios in cardiac tissues. CM mutations in the metavinculin tail domain (MVt) occur within the extra 68-residue insert that differentiates it from the vinculin tail domain (Vt). Vt binds actin filaments (F-actin) and promotes vinculin dimerization to bundle F-actin into thick fibers. While MVt binds to F-actin in a similar manner to Vt, MVt is incapable of F-actin bundling and inhibits Vt-mediated F-actin bundling. We performed F-actin co-sedimentation and negative-stain EM experiments to dissect the coordinated roles of metavinculin and vinculin in actin fiber assembly and the effects of three known metavinculin CM mutations. These CM mutants were found to weakly induce the formation of disordered F-actin assemblies. Notably, they fail to inhibit Vt-mediated F-actin bundling and instead promote formation of large assemblies embedded with linear bundles. Computational models of MVt bound to F-actin suggest that MVt undergoes a conformational change licensing the formation of a protruding sub-domain incorporating the insert, which sterically prevents dimerization and bundling of F-actin by Vt. Sub-domain formation is destabilized by CM mutations, disrupting this inhibitory mechanism. These findings provide new mechanistic insights into the ability of metavinculin to tune actin organization by vinculin and suggest that dysregulation of this process by CM mutants could underlie their malfunction in disease.


Assuntos
Actinas/metabolismo , Cardiomiopatias/genética , Mutação Puntual , Vinculina/genética , Animais , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Galinhas , Humanos , Modelos Moleculares , Ligação Proteica , Domínios Proteicos , Mapas de Interação de Proteínas , Vinculina/química , Vinculina/metabolismo
9.
PLoS Pathog ; 15(2): e1007536, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30716130

RESUMO

Extracellular signaling is a mechanism that higher eukaryotes have evolved to facilitate organismal homeostasis. Recent years have seen an emerging interest in the role of secreted microvesicles, termed extracellular vesicles (EV) or exosomes in this signaling network. EV contents can be modified by the cell in response to stimuli, allowing them to relay information to neighboring cells, influencing their physiology. Here we show that the tumor virus Kaposi's Sarcoma-associated herpesvirus (KSHV) hijacks this signaling pathway to induce cell proliferation, migration, and transcriptome reprogramming in cells not infected with the virus. KSHV-EV activates the canonical MEK/ERK pathway, while not alerting innate immune regulators, allowing the virus to exert these changes without cellular pathogen recognition. Collectively, we propose that KSHV establishes a niche favorable for viral spread and cell transformation through cell-derived vesicles, all while avoiding detection.


Assuntos
Reprogramação Celular/fisiologia , Vesículas Extracelulares/fisiologia , Herpesvirus Humano 8/metabolismo , Movimento Celular , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Reprogramação Celular/genética , Células Endoteliais/fisiologia , Herpesvirus Humano 8/genética , Interações Hospedeiro-Patógeno , Células Endoteliais da Veia Umbilical Humana , Humanos , Linfoma/genética , Linfoma/metabolismo , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virologia , Transdução de Sinais , Transcriptoma/genética , Proteínas Virais , Latência Viral
10.
J Extracell Vesicles ; 7(1): 1541396, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30533204

RESUMO

Isolation of extracellular vesicles (EVs) from cell culture supernatant or plasma can be accomplished in a variety of ways. Common measures to quantify relative success are: concentration of the EVs, purity from non-EVs associated protein, size homogeneity and functionality of the final product. Here, we present an industrial-scale workflow for isolating highly pure and functional EVs using cross-flow based filtration coupled with high-molecular weight Capto Core size exclusion. Through this combination, EVs loss is kept to a minimum. It outperforms other isolation procedures based on a number of biochemical and biophysical assays. Moreover, EVs isolated through this method can be further concentrated down or directly immunopurified to obtain discreet populations of EVs. From our results, we propose that cross-flow/Capto Core isolation is a robust method of purifying highly concentrated, homogenous, and functionally active EVs from industrial-scale input volumes with few contaminants relative to other methods.

11.
mBio ; 9(1)2018 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-29437924

RESUMO

Extracellular vesicles (EVs) or exosomes have been implicated in the pathophysiology of infections and cancer. The negative regulatory factor (Nef) encoded by simian immunodeficiency virus (SIV) and human immunodeficiency virus (HIV) plays a critical role in the progression to AIDS and impairs endosomal trafficking. Whether HIV-1 Nef can be loaded into EVs has been the subject of controversy, and nothing is known about the connection between SIV Nef and EVs. We find that both SIV and HIV-1 Nef proteins are present in affinity-purified EVs derived from cultured cells, as well as in EVs from SIV-infected macaques. Nef-positive EVs were functional, i.e., capable of membrane fusion and depositing their content into recipient cells. The EVs were able to transfer Nef into recipient cells. This suggests that Nef readily enters the exosome biogenesis pathway, whereas HIV virions are assembled at the plasma membrane. It suggests a novel mechanism by which lentiviruses can influence uninfected and uninfectable, i.e., CD4-negative, cells.IMPORTANCE Extracellular vesicles (EVs) transfer biologically active materials from one cell to another, either within the adjacent microenvironment or further removed. EVs also package viral RNAs, microRNAs, and proteins, which contributes to the pathophysiology of infection. In this report, we show that both human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) incorporate the virus-encoded Nef protein into EVs, including EVs circulating in the blood of SIV-infected macaques and that this presents a novel mechanism of Nef transfer to naive and even otherwise non-infectable cells. Nef is dispensable for viral replication but essential for AIDS progression in vivo Demonstrating that Nef incorporation into EVs is conserved across species implicates EVs as novel mediators of the pathophysiology of HIV. It could help explain the biological effects that HIV has on CD4-negative cells and EVs could become biomarkers of disease progression.


Assuntos
Exossomos/metabolismo , Produtos do Gene nef/metabolismo , HIV-1/fisiologia , Vírus da Imunodeficiência Símia/fisiologia , Animais , Células Cultivadas , Humanos , Macaca , Transporte Proteico
12.
mBio ; 8(5)2017 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-29018115

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) is associated with the human malignancy Kaposi's sarcoma and the lymphoproliferative disorders primary effusion lymphoma and multicentric Castleman's disease. KSHV establishes lytic infection of monocytes in vivo, which may represent an important cellular reservoir during KS disease progression. KS tumors consist of latently infected endothelial cells; however, lytic phase gene products are important for KS onset. Early KS lesion progression is driven by proinflammatory cytokines supplied by immune cell infiltrates including T cells and monocytes. KSHV-infected monocytes may supply the lytic viral products and the inflammatory milieu conducive to KS tumor progression. To establish successful infection, KSHV extensively modulates the host immune system. KSHV antigens activate both innate and adaptive immune responses including KSHV-specific T cells, but lifelong infection is still established. Programmed death ligand 1 (PD-L1) is a prosurvival cell surface protein that suppresses T-cell-mediated killing. PD-L1 is variably present on various tumor cells and is a targetable marker for cancer treatment. We show that KSHV infection of human monocytes increases PD-L1 expression and transcription in a dose-dependent manner. We also saw evidence of lytic gene expression in the KSHV-infected monocytes. Intact KSHV is needed for full PD-L1 response in human monocytes. KSHV induces a general proinflammatory cytokine milieu including interleukins 1α, 1ß, and 6, which have been implicated in early KS lesion progression. KSHV-mediated PD-L1 increase may represent a novel mechanism of KSHV-mediated immune modulation to allow for virus survival and eventually malignant progression.IMPORTANCE KSHV is the etiologic agent of Kaposi's sarcoma and the lymphoproliferative disorders primary effusion lymphoma and multicentric Castleman's disease. Programmed death ligand 1 (PD-L1) is an immunosuppressive cell surface marker that inhibits T cell activation. We report that KSHV infection of primary human monocytes upregulates PD-L1 transcription and protein expression. Analysis of the cytokine and chemokine milieu following KSHV infection of monocytes revealed that KSHV induces interleukins 1α, 1ß, and 6, all of which have been implicated in KS development. Our work has identified another potential immune evasion strategy for KSHV and a potential target for immunotherapy of KSHV-derived disease.


Assuntos
Antígeno B7-H1/genética , Citocinas/genética , Herpesvirus Humano 8/fisiologia , Interações Hospedeiro-Patógeno , Monócitos/imunologia , Monócitos/virologia , Citocinas/imunologia , Regulação Viral da Expressão Gênica , Humanos , Imunidade Inata , Regulação para Cima , Proteínas Virais/genética , Proteínas Virais/imunologia , Latência Viral
13.
J Virol ; 89(19): 9706-8, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26202231

RESUMO

Many longstanding questions about dynamics of virus-cell interactions can be answered by combining fluorescence imaging techniques with fluorescent protein (FP) tagging strategies. Successfully creating a FP fusion with a cellular or viral protein of interest first requires selecting the appropriate FP. However, while viral architecture and cellular localization often dictate the suitability of a FP, a FP's chemical and physical properties must also be considered. Here, we discuss the challenges of and offer suggestions for identifying the optimal FPs for studying the cell biology of viruses.


Assuntos
Microambiente Celular , Proteínas Luminescentes/metabolismo , Modelos Moleculares , Imagem Óptica/métodos , Proteínas Virais de Fusão/metabolismo , Virologia/métodos , Proteínas Luminescentes/química , Proteínas Virais de Fusão/química
14.
Nat Commun ; 6: 7670, 2015 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-26158227

RESUMO

To perform quantitative live cell imaging, investigators require fluorescent reporters that accurately report protein localization and levels, while minimally perturbing the cell. Yet, within the biochemically distinct environments of cellular organelles, popular fluorescent proteins (FPs), including EGFP, can be unreliable for quantitative imaging, resulting in the underestimation of protein levels and incorrect localization. Specifically, within the secretory pathway, significant populations of FPs misfold and fail to fluoresce due to non-native disulphide bond formation. Furthermore, transmembrane FP-fusion constructs can disrupt organelle architecture due to oligomerizing tendencies of numerous common FPs. Here, we describe a powerful set of bright and inert FPs optimized for use in multiple cellular compartments, especially oxidizing environments and biological membranes. Also, we provide new insights into the use of red FPs in the secretory pathway. Our monomeric 'oxFPs' finally resolve long-standing, underappreciated and important problems of cell biology and should be useful for a number of applications.


Assuntos
Membrana Celular/metabolismo , Corantes Fluorescentes/metabolismo , Proteínas Luminescentes/metabolismo , Animais , Proteínas de Bactérias/química , Linhagem Celular Tumoral , Cães , Corantes Fluorescentes/química , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Proteínas Luminescentes/química , Células Madin Darby de Rim Canino , Microscopia de Fluorescência , Imagem Óptica/métodos , Coloração e Rotulagem , Proteína Vermelha Fluorescente
15.
Virology ; 476: 240-248, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25555152

RESUMO

The HIV-1 envelope glycoprotein, gp120, binds the host cell receptor, CD4, in the initial step of HIV viral entry and infection. This process is an appealing target for the development of inhibitory drugs and neutralizing antibodies. To study gp120 binding and intracellular trafficking, we engineered a fluorescent fusion of the humanized gp120 JRFL HIV-1 variant and GFP. Gp120-sfGFP is glycosylated with human sugars, robustly expressed, and secreted from cultured human cells. Protein dynamics, quality control, and trafficking can be visualized in live cells. The fusion protein can be readily modified with different gp120 variants or fluorescent proteins. Finally, secreted gp120-sfGFP enables a sensitive and easy binding assay that can quantitatively screen potential inhibitors of gp120-CD4 binding on live cells via fluorescence imaging or laser scanning cytometry. This adaptable research tool should aid in studies of gp120 cell biology and the development of novel anti-HIV drugs.


Assuntos
Linfócitos T CD4-Positivos/virologia , Rastreamento de Células/métodos , Proteínas de Fluorescência Verde/metabolismo , Proteína gp120 do Envelope de HIV/metabolismo , Infecções por HIV/virologia , HIV-1/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Engenharia Genética , Proteínas de Fluorescência Verde/genética , Proteína gp120 do Envelope de HIV/genética , Infecções por HIV/metabolismo , HIV-1/genética , Humanos , Receptores Virais/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
16.
DNA Cell Biol ; 32(11): 622-7, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23971632

RESUMO

Fluorescent proteins (FPs) have been powerful tools for cell biologists for over 15 years. The large variety of FPs available rarely comes with an instruction manual or a warning label. The potential pitfalls of the use of FPs in cellular organelles represent a significant concern for investigators. FPs generally did not evolve in the often distinctive physicochemical environments of subcellular organelles. In organelles, FPs can misfold, go dark, and even distort organelle morphology. In this minireview, we describe the issues associated with FPs in organelles and provide solutions to enable investigators to better exploit FP technology in cells.


Assuntos
Proteínas Luminescentes/análise , Proteínas Luminescentes/química , Proteínas Luminescentes/metabolismo , Organelas/metabolismo , Animais , Dissulfetos , Fluorescência , Glicosilação , Humanos , Proteínas Luminescentes/genética , Biologia Molecular/métodos , Oxirredução , Conformação Proteica , Proteínas Recombinantes/análise , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
17.
Biochem Biophys Res Commun ; 430(3): 1114-9, 2013 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-23257162

RESUMO

Fluorescent protein (FP) technologies suitable for use within the eukaryotic secretory pathway are essential for live cell and protein dynamic studies. Localization of FPs within the endoplasmic reticulum (ER) lumen has potentially significant consequences for FP function. All FPs are resident cytoplasmic proteins and have rarely been evolved for the chemically distinct environment of the ER lumen. In contrast to the cytoplasm, the ER lumen is oxidizing and the site where secretory proteins are post-translationally modified by disulfide bond formation and N-glycosylation on select asparagine residues. Cysteine residues and N-linked glycosylation consensus sequences were identified within many commonly utilized FPs. Here, we report mTagBFP is post-translationally modified when localized to the ER lumen. Our findings suggest these modifications can grossly affect the sensitivity and reliability of FP tools within the secretory pathway. To optimize tools for studying events in this important intracellular environment, we modified mTagBFP by mutating its cysteines and consensus N-glycosylation sites. We report successful creation of a secretory pathway-optimized blue FP, secBFP2.


Assuntos
Cisteína/química , Células Eucarióticas/metabolismo , Proteínas Luminescentes/química , Via Secretória , Sequência de Aminoácidos , Substituição de Aminoácidos , Linhagem Celular Tumoral , Cisteína/genética , Retículo Endoplasmático/metabolismo , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Dados de Sequência Molecular , Mutagênese , Engenharia de Proteínas , Dobramento de Proteína , Processamento de Proteína Pós-Traducional
18.
Traffic ; 13(5): 643-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22289035

RESUMO

Several fluorescent proteins (FPs) are prone to forming low-affinity oligomers. This undesirable tendency is exacerbated when FPs are confined to membranes or when fused to naturally oligomeric proteins. Oligomerization of FPs limits their suitability for creating fusions with proteins of interest. Unfortunately, no standardized method evaluates the biologically relevant oligomeric state of FPs. Here, we describe a quantitative visual assay for assessing whether FPs are sufficiently monomeric under physiologic conditions. Membrane-associated FP-fusion proteins, by virtue of their constrained planar geometry, achieve high effective concentrations. We exploited this propensity to develop an assay to measure FP tendencies to oligomerize in cells. FPs were fused on the cytoplasmic end of an endoplasmic reticulum (ER) signal-anchor membrane protein (CytERM) and expressed in cells. Cells were scored based on the ability of CytERM to homo-oligomerize with proteins on apposing membranes and restructure the ER from a tubular network into organized smooth ER (OSER) whorl structures. The ratio of nuclear envelope and OSER structures mean fluorescent intensities for cells expressing enhanced green fluorescent protein (EGFP) or monomeric green fluorescent protein (mGFP) CytERM established standards for comparison of uncharacterized FPs. We tested three FPs and identified two as sufficiently monomeric, while a third previously reported as monomeric was found to strongly oligomerize.


Assuntos
Proteínas de Fluorescência Verde/química , Animais , Sistema Enzimático do Citocromo P-450/química , Citoplasma/metabolismo , Citosol/metabolismo , Retículo Endoplasmático/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Immunoblotting/métodos , Microscopia Eletrônica/métodos , Membrana Nuclear/metabolismo , Osteossarcoma/metabolismo , Plasmídeos/metabolismo , Proteínas Recombinantes de Fusão/química , Transdução de Sinais
19.
Alcohol Clin Exp Res ; 36(1): 14-23, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21790674

RESUMO

BACKGROUND: Many alcoholic patients have serum protein deficiency that contributes to their systemic problems. The unfolded protein response (UPR) is induced in response to disequilibrium in the protein folding capability of the endoplasmic reticulum (ER) and is implicated in hepatocyte lipid accumulation and apoptosis, which are associated with alcoholic liver disease (ALD). We investigated whether alcohol affects ER structure, function, and UPR activation in hepatocytes in vitro and in vivo. METHODS: HepG2 cells expressing human cytochrome P450 2E1 and mouse alcohol dehydrogenase (VL-17A) were treated for up to 48 hours with 50 and 100 mM ethanol. Zebrafish larvae at 4 days postfertilization were exposed to 350 mM ethanol for 32 hours. ER morphology was visualized by fluorescence in cells and transmission electron microscopy in zebrafish. UPR target gene activation was assessed using quantitative PCR, in situ hybridization, and Western blotting. Mobility of the major ER chaperone, BIP, was monitored in cells by fluorescence recovery after photobleaching (FRAP). RESULTS: VL-17A cells metabolized alcohol yet only had slight activation of some UPR target genes following ethanol treatment. However, ER fragmentation, crowding, and accumulation of unfolded proteins as detected by immunofluorescence and FRAP demonstrate that alcohol induced some ER dysfunction despite the lack of UPR activation. Zebrafish treated with alcohol, however, showed modest ER dilation, and several UPR targets were significantly induced. CONCLUSIONS: Ethanol metabolism directly impairs ER structure and function in hepatocytes. Zebrafish are a novel in vivo system for studying ALD.


Assuntos
Retículo Endoplasmático/efeitos dos fármacos , Etanol/toxicidade , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Animais , Retículo Endoplasmático/ultraestrutura , Células Hep G2 , Humanos , Camundongos , Peixe-Zebra
20.
PLoS One ; 6(2): e14647, 2011 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-21311600

RESUMO

BACKGROUND: Inhalation of crystalline silica is known to cause an inflammatory reaction and chronic exposure leads to lung fibrosis and can progress into the disease, silicosis. Cultured macrophages bind crystalline silica particles, phagocytose them, and rapidly undergo apoptotic and necrotic death. The mechanism by which particles are bound and internalized and the reason particles are toxic is unclear. Amorphous silica has been considered to be a less toxic form, but this view is controversial. We compared the uptake and toxicity of amorphous silica to crystalline silica. METHODOLOGY/PRINCIPAL FINDINGS: Amorphous silica particles are phagocytosed by macrophage cells and a single internalized particle is capable of killing a cell. Fluorescent dextran is released from endo-lysosomes within two hours after silica treatment and Caspase-3 activation occurs within 4 hours. Interestingly, toxicity is specific to macrophage cell lines. Other cell types are resistant to silica particle toxicity even though they internalize the particles. The large and uniform size of the spherical, amorphous silica particles allowed us to monitor them during the uptake process. In mCherry-actin transfected macrophages, actin rings began to form 1-3 minutes after silica binding and the actin coat disassembled rapidly following particle internalization. Pre-loading cells with fluorescent dextran allowed us to visualize the fusion of phagosomes with endosomes during internalization. These markers provided two new ways to visualize and quantify particle internalization. At 37 °C the rate of amorphous silica internalization was very rapid regardless of particle coating. However, at room temperature, opsonized silica is internalized much faster than non-opsonized silica. CONCLUSIONS/SIGNIFICANCE: Our results indicate that amorphous and crystalline silica are both phagocytosed and both toxic to mouse alveolar macrophage (MH-S) cells. The pathway leading to apoptosis appears to be similar in both cases. However, the result suggests a mechanistic difference between FcγRIIA receptor-mediated and non-opsonized silica particle phagocytosis.


Assuntos
Fagocitose/fisiologia , Dióxido de Silício/toxicidade , Animais , Células COS , Morte Celular/efeitos dos fármacos , Células Cultivadas , Chlorocebus aethiops , Cristalização , Cães , Células HeLa , Humanos , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patologia , Macrófagos Alveolares/fisiologia , Camundongos , Células NIH 3T3 , Ratos , Silicose/imunologia , Silicose/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...