Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Breast Cancer Res ; 26(1): 78, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38750591

RESUMO

BACKGROUND: Metastatic breast cancer is a leading cause of cancer death in woman. Current treatment options are often associated with adverse side effects and poor outcomes, demonstrating the need for effective new treatments. Immunotherapies can provide durable outcomes in many cancers; however, limited success has been achieved in metastatic triple negative breast cancer. We tested whether combining different immunotherapies can target metastatic triple negative breast cancer in pre-clinical models. METHODS: Using primary and metastatic 4T1 triple negative mammary carcinoma models, we examined the therapeutic effects of oncolytic vesicular stomatitis virus (VSVΔM51) engineered to express reovirus-derived fusion associated small transmembrane proteins p14 (VSV-p14) or p15 (VSV-p15). These viruses were delivered alone or in combination with natural killer T (NKT) cell activation therapy mediated by adoptive transfer of α-galactosylceramide-loaded dendritic cells. RESULTS: Treatment of primary 4T1 tumors with VSV-p14 or VSV-p15 alone increased immunogenic tumor cell death, attenuated tumor growth, and enhanced immune cell infiltration and activation compared to control oncolytic virus (VSV-GFP) treatments and untreated mice. When combined with NKT cell activation therapy, oncolytic VSV-p14 and VSV-p15 reduced metastatic lung burden to undetectable levels in all mice and generated immune memory as evidenced by enhanced in vitro recall responses (tumor killing and cytokine production) and impaired tumor growth upon rechallenge. CONCLUSION: Combining NKT cell immunotherapy with enhanced oncolytic virotherapy increased anti-tumor immune targeting of lung metastasis and presents a promising treatment strategy for metastatic breast cancer.


Assuntos
Células T Matadoras Naturais , Terapia Viral Oncolítica , Vírus Oncolíticos , Animais , Feminino , Camundongos , Células T Matadoras Naturais/imunologia , Terapia Viral Oncolítica/métodos , Humanos , Linhagem Celular Tumoral , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Imunoterapia/métodos , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular Indiana/imunologia , Neoplasias de Mama Triplo Negativas/terapia , Neoplasias de Mama Triplo Negativas/imunologia , Neoplasias de Mama Triplo Negativas/patologia , Terapia Combinada , Metástase Neoplásica , Vesiculovirus/genética , Células Dendríticas/imunologia , Neoplasias da Mama/terapia , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Modelos Animais de Doenças
2.
Viruses ; 12(7)2020 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-32610593

RESUMO

The Reoviridae family is the only non-enveloped virus family with members that use syncytium formation to promote cell-cell virus transmission. Syncytiogenesis is mediated by a fusion-associated small transmembrane (FAST) protein, a novel family of viral membrane fusion proteins. Previous evidence suggested the fusogenic reoviruses arose from an ancestral non-fusogenic virus, with the preponderance of fusogenic species suggesting positive evolutionary pressure to acquire and maintain the fusion phenotype. New phylogenetic analyses that included the atypical waterfowl subgroup of avian reoviruses and recently identified new orthoreovirus species indicate a more complex relationship between reovirus speciation and fusogenic capacity, with numerous predicted internal indels and 5'-terminal extensions driving the evolution of the orthoreovirus' polycistronic genome segments and their encoded FAST and fiber proteins. These inferred recombination events generated bi- and tricistronic genome segments with diverse gene constellations, they occurred pre- and post-orthoreovirus speciation, and they directly contributed to the evolution of the four extant orthoreovirus FAST proteins by driving both the gain and loss of fusion capability. We further show that two distinct post-speciation genetic events led to the loss of fusion in the waterfowl isolates of avian reovirus, a recombination event that replaced the p10 FAST protein with a heterologous, non-fusogenic protein and point substitutions in a conserved motif that destroyed the p10 assembly into multimeric fusion platforms.


Assuntos
Doenças das Aves/virologia , Evolução Molecular , Genoma Viral , Orthoreovirus/genética , Infecções por Reoviridae/veterinária , Proteínas Virais de Fusão/metabolismo , Sequência de Aminoácidos , Animais , Animais Selvagens/virologia , Anseriformes/virologia , Mutação com Ganho de Função , Especiação Genética , Células Gigantes/virologia , Orthoreovirus/classificação , Orthoreovirus/isolamento & purificação , Orthoreovirus/fisiologia , Filogenia , Infecções por Reoviridae/virologia , Alinhamento de Sequência , Proteínas Virais de Fusão/genética
3.
Annu Rev Virol ; 6(1): 341-363, 2019 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-31283438

RESUMO

With no limiting membrane surrounding virions, nonenveloped viruses have no need for membrane fusion to gain access to intracellular replication compartments. Consequently, nonenveloped viruses do not encode membrane fusion proteins. The only exception to this dogma is the fusogenic reoviruses that encode fusion-associated small transmembrane (FAST) proteins that induce syncytium formation. FAST proteins are the smallest viral membrane fusion proteins and, unlike their enveloped virus counterparts, are nonstructural proteins that evolved specifically to induce cell-to-cell, not virus-cell, membrane fusion. This distinct evolutionary imperative is reflected in structural and functional features that distinguish this singular family of viral fusogens from all other protein fusogens. These rudimentary fusogens comprise specific combinations of different membrane effector motifs assembled into small, modular membrane fusogens. FAST proteins offer a minimalist model to better understand the ubiquitous process of protein-mediated membrane fusion and to reveal novel mechanisms of nonenveloped virus dissemination that contribute to virulence.


Assuntos
Reoviridae/metabolismo , Proteínas Virais de Fusão/metabolismo , Sequência de Aminoácidos , Animais , Doenças dos Peixes/virologia , Humanos , Filogenia , Reoviridae/classificação , Reoviridae/genética , Infecções por Reoviridae/veterinária , Infecções por Reoviridae/virologia , Alinhamento de Sequência , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/genética , Internalização do Vírus
4.
Autophagy ; 15(4): 686-706, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30444165

RESUMO

Cancer stem-like cells (CSCs), a small population of pluripotent cells residing within heterogeneous tumor mass, remain highly resistant to various chemotherapies as compared to the differentiated cancer cells. It is being postulated that CSCs possess unique molecular mechanisms, such as autophagic homeostasis, that allow CSCs to withstand the therapeutic assaults. Here we demonstrate that HDAC6 inhibition differentially modulates macroautophagy/autophagy in CSCs as compared to that of differentiated cancer cells. Using human and murine CSC models and differentiated cells, we show that the inhibition or knockdown (KD) of HDAC6 decreases CSC pluripotency by downregulating major pluripotency factors POU5F1, NANOG and SOX2. This decreased HDAC6 expression increases ACTB, TUBB3 and CSN2 expression and promotes differentiation in CSCs in an apoptosis-independent manner. Mechanistically, HDAC6 KD in CSCs decreases pluripotency by promoting autophagy, whereas the inhibition of pluripotency via retinoic acid treatment, POU5F1 or autophagy-related gene (ATG7 and ATG12) KD in CSCs decreases HDAC6 expression and promotes differentiation. Interestingly, HDAC6 KD-mediated CSC growth inhibition is further enhanced in the presence of autophagy inducers Tat-Beclin 1 peptide and rapamycin. In contrast to the results observed in CSCs, HDAC6 KD in differentiated breast cancer cells downregulates autophagy and increases apoptosis. Furthermore, the autophagy regulator p-MTOR, upstream negative regulators of p-MTOR (TSC1 and TSC2) and downstream effectors of p-MTOR (p-RPS6KB and p-EIF4EBP1) are differentially regulated in CSCs versus differentiated cancer cells following HDAC6 KD. Overall these data identify the differential regulation of autophagy as a molecular link behind the differing chemo-susceptibility of CSCs and differentiated cancer cells.


Assuntos
Autofagia/genética , Neoplasias da Mama/metabolismo , Diferenciação Celular/genética , Desacetilase 6 de Histona/antagonistas & inibidores , Desacetilase 6 de Histona/metabolismo , Células-Tronco Neoplásicas/metabolismo , Actinas/metabolismo , Animais , Apoptose/genética , Proteína 12 Relacionada à Autofagia/genética , Proteína 12 Relacionada à Autofagia/metabolismo , Proteína 7 Relacionada à Autofagia/genética , Proteína 7 Relacionada à Autofagia/metabolismo , Neoplasias da Mama/genética , Sobrevivência Celular/genética , Feminino , Células HEK293 , Desacetilase 6 de Histona/genética , Humanos , Camundongos , Proteína Homeobox Nanog/genética , Proteína Homeobox Nanog/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Proteoma/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteína 1 do Complexo Esclerose Tuberosa/antagonistas & inibidores , Proteína 1 do Complexo Esclerose Tuberosa/genética , Proteína 1 do Complexo Esclerose Tuberosa/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa/antagonistas & inibidores , Proteína 2 do Complexo Esclerose Tuberosa/genética , Proteína 2 do Complexo Esclerose Tuberosa/metabolismo
5.
Dev Cell ; 46(6): 676-678, 2018 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-30253167

RESUMO

Muscle-forming myotubes are formed from the cell-cell fusion of myoblasts. In this issue of Developmental Cell, Leikina et al. (2018) provide compelling evidence that myoblast fusion is dependent on a two-component membrane fusion complex of Myomaker and Myomerger, which function in hemifusion and pore formation activity, respectively.


Assuntos
Desenvolvimento Muscular , Proteínas Musculares , Fusão Celular , Proteínas de Membrana , Mioblastos
6.
Mol Ther Oncolytics ; 6: 80-89, 2017 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-28856238

RESUMO

The reovirus fusion-associated small transmembrane (FAST) proteins are the smallest known viral fusogens (∼100-150 amino acids) and efficiently induce cell-cell fusion and syncytium formation in multiple cell types. Syncytium formation enhances cell-cell virus transmission and may also induce immunogenic cell death, a form of apoptosis that stimulates immune recognition of tumor cells. These properties suggest that FAST proteins might serve to enhance oncolytic virotherapy. The oncolytic activity of recombinant VSVΔM51 (an interferon-sensitive vesicular stomatitis virus [VSV] mutant) encoding the p14 FAST protein (VSV-p14) was compared with a similar construct encoding GFP (VSV-GFP) in cell culture and syngeneic BALB/c tumor models. Compared with VSV-GFP, VSV-p14 exhibited increased oncolytic activity against MCF-7 and 4T1 breast cancer spheroids in culture and reduced primary 4T1 breast tumor growth in vivo. VSV-p14 prolonged survival in both primary and metastatic 4T1 breast cancer models, and in a CT26 metastatic colon cancer model. As with VSV-GFP, VSV-p14 preferentially replicated in vivo in tumors and was cleared rapidly from other sites. Furthermore, VSV-p14 increased the numbers of activated splenic CD4, CD8, natural killer (NK), and natural killer T (NKT) cells, and increased the number of activated CD4 and CD8 cells in tumors. FAST proteins may therefore provide a multi-pronged approach to improving oncolytic virotherapy via syncytium formation and enhanced immune stimulation.

7.
Cell Death Dis ; 8(3): e2724, 2017 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-28358373

RESUMO

The promyelocytic leukemia protein (PML) is expressed in most normal human tissues and forms nuclear bodies (NBs) that have roles in gene regulation and cellular processes such as DNA repair, cell cycle control, and cell fate decisions. Using murine C2C12 myoblasts, we demonstrate that activation of skeletal muscle differentiation results in loss of PML and PML NBs prior to myotube fusion. Myotube formation was associated with marked chromatin reorganization and the relocalization of DAXX from PML NBs to chromocentres. MyoD expression was sufficient to cause PML NB loss, and silencing of PML induced DAXX relocalization. Fusion of C2C12 cells using the reptilian reovirus p14 fusogenic protein failed to disrupt PML NBs yet still promoted DAXX redistribution and loss; whereas ectopic expression of PML in differentiated cells only partially restored PML NB formation and DAXX localization at NBs. Finally, we determined that the C-terminal SUMO-interacting motif of DAXX is required for its colocalization with ATRX in heterochromatin domains during myotube formation. These data support a model in which activation of myogenic differentiation results in PML NB loss, chromatin reorganization and DAXX relocalization, and provides a paradigm for understanding the consequence of PML loss in other cellular contexts, such as during cancer development and progression.


Assuntos
Proteínas de Transporte/metabolismo , Heterocromatina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Modelos Biológicos , Desenvolvimento Muscular/fisiologia , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/metabolismo , Proteínas Nucleares/metabolismo , Proteína da Leucemia Promielocítica/metabolismo , Motivos de Aminoácidos , Animais , Proteínas de Transporte/genética , Linhagem Celular , Proteínas Correpressoras , Heterocromatina/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Chaperonas Moleculares , Fibras Musculares Esqueléticas/citologia , Mioblastos/citologia , Proteínas Nucleares/genética , Proteína da Leucemia Promielocítica/genética , Transporte Proteico/fisiologia
8.
Mol Oncol ; 10(9): 1485-1496, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27592281

RESUMO

Breast cancer stem cells (CSCs) can be identified by increased Aldefluor fluorescence caused by increased expression of aldehyde dehydrogenase 1A3 (ALDH1A3), as well as ALDH1A1 and ALDH2. In addition to being a CSC marker, ALDH1A3 regulates gene expression via retinoic acid (RA) signaling and plays a key role in the progression and chemotherapy resistance of cancer. Therefore, ALDH1A3 represents a druggable anti-cancer target of interest. Since to date, there are no characterized ALDH1A3 isoform inhibitors, drugs that were previously described as inhibiting the activity of other ALDH isoforms were tested for anti-ALDH1A3 activity. Twelve drugs (3-hydroxy-dl-kynurenine, benomyl, citral, chloral hydrate, cyanamide, daidzin, DEAB, disulfiram, gossypol, kynurenic acid, molinate, and pargyline) were compared for their efficacy in inducing apoptosis and reducing ALDH1A3, ALDH1A1 and ALDH2-associated Aldefluor fluorescence in breast cancer cells. Citral was identified as the best inhibitor of ALDH1A3, reducing the Aldefluor fluorescence in breast cancer cell lines and in a patient-derived tumor xenograft. Nanoparticle encapsulated citral specifically reduced the enhanced tumor growth of MDA-MB-231 cells overexpressing ALDH1A3. To determine the potential mechanisms of citral-mediated tumor growth inhibition, we performed cell proliferation, clonogenic, and gene expression assays. Citral reduced ALDH1A3-mediated colony formation and expression of ALDH1A3-inducible genes. In conclusion, citral is an effective ALDH1A3 inhibitor and is able to block ALDH1A3-mediated breast tumor growth, potentially via blocking its colony forming and gene expression regulation activity. The promise of ALDH1A3 inhibitors as adjuvant therapies for patients with tumors that have a large population of high-ALDH1A3 CSCs is discussed.


Assuntos
Aldeído Oxirredutases/metabolismo , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Monoterpenos/uso terapêutico , Células-Tronco Neoplásicas/metabolismo , Monoterpenos Acíclicos , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Fluorescência , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos Endogâmicos NOD , Camundongos SCID , Monoterpenos/química , Monoterpenos/farmacologia , Nanopartículas/química , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Ensaio Tumoral de Célula-Tronco , Ensaios Antitumorais Modelo de Xenoenxerto
9.
PLoS Pathog ; 11(6): e1004962, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26061049

RESUMO

Pore formation is the most energy-demanding step during virus-induced membrane fusion, where high curvature of the fusion pore rim increases the spacing between lipid headgroups, exposing the hydrophobic interior of the membrane to water. How protein fusogens breach this thermodynamic barrier to pore formation is unclear. We identified a novel fusion-inducing lipid packing sensor (FLiPS) in the cytosolic endodomain of the baboon reovirus p15 fusion-associated small transmembrane (FAST) protein that is essential for pore formation during cell-cell fusion and syncytiogenesis. NMR spectroscopy and mutational studies indicate the dependence of this FLiPS on a hydrophobic helix-loop-helix structure. Biochemical and biophysical assays reveal the p15 FLiPS preferentially partitions into membranes with high positive curvature, and this partitioning is impeded by bis-ANS, a small molecule that inserts into hydrophobic defects in membranes. Most notably, the p15 FLiPS can be functionally replaced by heterologous amphipathic lipid packing sensors (ALPS) but not by other membrane-interactive amphipathic helices. Furthermore, a previously unrecognized amphipathic helix in the cytosolic domain of the reptilian reovirus p14 FAST protein can functionally replace the p15 FLiPS, and is itself replaceable by a heterologous ALPS motif. Anchored near the cytoplasmic leaflet by the FAST protein transmembrane domain, the FLiPS is perfectly positioned to insert into hydrophobic defects that begin to appear in the highly curved rim of nascent fusion pores, thereby lowering the energy barrier to stable pore formation.


Assuntos
Fusão de Membrana/fisiologia , Reoviridae/fisiologia , Proteínas Virais de Fusão/metabolismo , Animais , Chlorocebus aethiops , Cromatografia Líquida de Alta Pressão , Dicroísmo Circular , Espectroscopia de Ressonância Magnética , Conformação Proteica , Reoviridae/patogenicidade , Transfecção , Células Vero , Proteínas Virais de Fusão/química
10.
Oncotarget ; 6(13): 11162-74, 2015 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-25883213

RESUMO

Synaptopodin-2 (Synpo2), an actin-binding protein and invasive cancer biomarker, induces formation of complex stress fiber networks in the cell body and promotes PC3 prostate cancer cell migration in response to serum stimulation. The role of these actin networks in enhanced cancer cell migration is unknown. Using time-course analysis and live cell imaging of mock- and Synpo2-transduced PC3 cells, we now show that Synpo2 induces assembly of actin fibers near the cell periphery and Arp2/3-dependent lamellipodia formation. Lamellipodia formed in a non-directional manner or repeatedly changed direction, explaining the enhanced chemokinetic activity of PC3 cells in response to serum stimulation. Myosin contraction promotes retrograde flow of the Synpo2-associated actin filaments at the leading edge and their merger with actin networks in the cell body. Enhanced PC3 cell migration correlates with Synpo2-induced formation of lamellipodia and immature focal adhesions (FAs), but is not dependent on myosin contraction or FA maturation. The previously reported correlation between Synpo2-induced stress fiber assembly and enhanced PC3 cell migration therefore reflects the role of Synpo2 as a newly identified regulator of actin bundle formation and nascent FA assembly near the leading cell edge.


Assuntos
Citoesqueleto de Actina/metabolismo , Movimento Celular , Citoesqueleto/metabolismo , Adesões Focais/fisiologia , Proteínas dos Microfilamentos/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Pseudópodes/patologia , Proteína 2 Relacionada a Actina/metabolismo , Proteína 3 Relacionada a Actina/metabolismo , Apoptose , Western Blotting , Proliferação de Células , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Masculino , Microscopia de Fluorescência , Cadeias Leves de Miosina/antagonistas & inibidores , Cadeias Leves de Miosina/genética , Cadeias Leves de Miosina/metabolismo , Pseudópodes/metabolismo , RNA Interferente Pequeno/genética , Células Tumorais Cultivadas
11.
Am J Pathol ; 185(4): 927-42, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25794704

RESUMO

Macrophages are increasingly recognized as a potential therapeutic target in myocardial fibrosis via interactions with fibroblasts. We have characterized macrophage depletion and inhibition of nonclassical macrophage migration, in addition to direct interactions between nonclassical macrophages and fibroblasts in angiotensin II (AngII)-mediated, hypertensive myocardial fibrosis. Macrophage depletion was achieved by daily i.v. clodronate liposomes (-1 day to +3 days) during AngII infusion. Cx3cr1(-/-) mice were used to inhibit nonclassical macrophage migration. Macrophage phenotype (F4/80, CD11b, Ly6C) was characterized by immunofluorescence and flow cytometry. Collagen was assessed by Sirius Red/Fast Green. Quantitative real-time RT-PCR was performed for transcript levels. AngII/wild-type (WT) mice displayed significant infiltrate and fibrosis compared with saline/WT, which was virtually ablated by clodronate liposomes independent of hypertension. In vitro data supported M2 macrophages promoting fibroblast differentiation and collagen production. AngII/Cx3cr1(-/-) mice, however, significantly increased macrophage infiltrate and fibrosis relative to AngII/WT. AngII/Cx3cr1(-/-) mice also showed an M1 phenotypic shift relative to WT mice in, which the predominant phenotype was Ly6C(low), CD206(+) (M2). Myocardial IL-1ß was significantly up-regulated, whereas transforming growth factor ß down-regulated with this M1 shift. We demonstrated that infiltrating macrophages are critical to AngII-mediated myocardial fibrosis by preventing the development of fibrosis after liposomal depletion of circulating monocytes. Our findings also suggest that some macrophages, namely M2, may confer a protective myocardial environment that may prevent excessive tissue injury.


Assuntos
Macrófagos/metabolismo , Miocárdio/patologia , Actinas/metabolismo , Administração Intravenosa , Angiotensina II/administração & dosagem , Angiotensina II/farmacologia , Animais , Antígenos Ly/metabolismo , Receptor 1 de Quimiocina CX3C , Ácido Clodrônico/administração & dosagem , Ácido Clodrônico/farmacologia , Colágeno/biossíntese , Eletrocardiografia , Fibrose , Mediadores da Inflamação/metabolismo , Lipossomos/administração & dosagem , Lipossomos/farmacologia , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Miocárdio/metabolismo , Células NIH 3T3 , Receptores de Quimiocinas/deficiência , Receptores de Quimiocinas/metabolismo
12.
Biochim Biophys Acta ; 1848(2): 408-16, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25450808

RESUMO

The reovirus p10 fusion-associated small transmembrane (FAST) proteins are the smallest known membrane fusion proteins, and evolved specifically to mediate cell-cell, rather than virus-cell, membrane fusion. The 36-40-residue ectodomains of avian reovirus (ARV) and Nelson Bay reovirus (NBV) p10 contain an essential intramolecular disulfide bond required for both cell-cell fusion and lipid mixing between liposomes. To more clearly define the functional, biochemical and biophysical features of this novel fusion peptide, synthetic peptides representing the p10 ectodomains of ARV and NBV were analyzed by solution-state NMR spectroscopy, circular dichroism spectroscopy, fluorescence spectroscopy-based hydrophobicity analysis, and liposome binding and fusion assays. Results indicate that disulfide bond formation promotes exposure of hydrophobic residues, as indicated by bis-ANS binding and time-dependent peptide aggregation under aqueous conditions, implying the disulfide bond creates a small, geometrically constrained, cystine noose. Noose formation is required for peptide partitioning into liposome membranes and liposome lipid mixing, and electron microscopy revealed that liposome-liposome fusion occurs in the absence of liposome tubulation. In addition, p10 fusion peptide activity, but not membrane partitioning, is dependent on membrane cholesterol.


Assuntos
Colesterol/química , Cistina/química , Lipossomos/química , Proteínas Virais de Fusão/química , Sequência de Aminoácidos , Interações Hidrofóbicas e Hidrofílicas , Fusão de Membrana , Dados de Sequência Molecular , Orthoreovirus/química , Orthoreovirus Aviário/química , Estrutura Secundária de Proteína , Proteínas Virais de Fusão/síntese química
13.
Trends Microbiol ; 22(12): 715-24, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25245455

RESUMO

Reovirus fusion-associated small transmembrane (FAST) proteins are the only known nonenveloped virus fusogens and are dedicated to inducing cell-to-cell, not virus-cell, membrane fusion. Numerous structural and functional attributes distinguish this novel family of viral fusogens from all enveloped virus membrane fusion proteins. Both families of viral fusogens play key roles in virus dissemination and pathogenicity, but employ different mechanisms to mediate membrane apposition and merger. However, convergence of these distinct families of viral membrane fusion proteins on common pathways needed for pore expansion and syncytium formation suggests syncytiogenesis represents a cellular response to the presence of cell-cell fusion pores. Together, FAST proteins and enveloped virus fusion proteins provide exceptional insights into the ubiquitous process of cell-cell membrane fusion and syncytium formation.


Assuntos
Células Gigantes/fisiologia , Fusão de Membrana , Reoviridae/metabolismo , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo , Sequência de Aminoácidos , Dados de Sequência Molecular , Orthoreovirus/metabolismo , Filogenia , Alinhamento de Sequência , Internalização do Vírus
14.
PLoS Pathog ; 10(3): e1004023, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24651689

RESUMO

The homologous p10 fusion-associated small transmembrane (FAST) proteins of the avian (ARV) and Nelson Bay (NBV) reoviruses are the smallest known viral membrane fusion proteins, and are virulence determinants of the fusogenic reoviruses. The small size of FAST proteins is incompatible with the paradigmatic membrane fusion pathway proposed for enveloped viral fusion proteins. Understanding how these diminutive viral fusogens mediate the complex process of membrane fusion is therefore of considerable interest, from both the pathogenesis and mechanism-of-action perspectives. Using chimeric ARV/NBV p10 constructs, the 36-40-residue ectodomain was identified as the major determinant of the differing fusion efficiencies of these homologous p10 proteins. Extensive mutagenic analysis determined the ectodomain comprises two distinct, essential functional motifs. Syncytiogenesis assays, thiol-specific surface biotinylation, and liposome lipid mixing assays identified an ∼25-residue, N-terminal motif that dictates formation of a cystine loop fusion peptide in both ARV and NBV p10. Surface immunofluorescence staining, FRET analysis and cholesterol depletion/repletion studies determined the cystine loop motif is connected through a two-residue linker to a 13-residue membrane-proximal ectodomain region (MPER). The MPER constitutes a second, independent motif governing reversible, cholesterol-dependent assembly of p10 multimers in the plasma membrane. Results further indicate that: (1) ARV and NBV homomultimers segregate to distinct, cholesterol-dependent microdomains in the plasma membrane; (2) p10 homomultimerization and cholesterol-dependent microdomain localization are co-dependent; and (3) the four juxtamembrane MPER residues present in the multimerization motif dictate species-specific microdomain association and homomultimerization. The p10 ectodomain therefore constitutes a remarkably compact, multifunctional fusion module that directs syncytiogenic efficiency and species-specific assembly of p10 homomultimers into cholesterol-dependent fusion platforms in the plasma membrane.


Assuntos
Orthoreovirus/fisiologia , Infecções por Reoviridae/metabolismo , Proteínas Virais de Fusão/metabolismo , Internalização do Vírus , Sequência de Aminoácidos , Animais , Western Blotting , Linhagem Celular , Colesterol/metabolismo , Citometria de Fluxo , Transferência Ressonante de Energia de Fluorescência , Microscopia de Fluorescência , Dados de Sequência Molecular , Multimerização Proteica , Estrutura Terciária de Proteína , Transfecção , Proteínas Virais de Fusão/química
15.
J Virol ; 88(11): 6528-31, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24672027

RESUMO

Using lysophosphatidylcholine, a curvature-inducing lysolipid, we have isolated a reversible, "stalled pore" phenotype during syncytium formation induced by the p14 fusion-associated small transmembrane (FAST) protein and influenza virus hemagglutinin (HA) fusogens. This is the first evidence that lateral propagation of stable fusion pores leading to syncytiogenesis mediated by diverse viral fusogens is inhibited by promotion of positive membrane curvature in the outer leaflets of the lipid bilayer surrounding intercellular fusion pores.


Assuntos
Células Gigantes/fisiologia , Células Gigantes/virologia , Hemaglutininas Virais/metabolismo , Lisofosfatidilcolinas/metabolismo , Fusão de Membrana/fisiologia , Modelos Biológicos , Proteínas Virais de Fusão/metabolismo , Animais , Chlorocebus aethiops , Microscopia de Fluorescência , Células Vero
16.
J Virol ; 88(11): 6137-47, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24648446

RESUMO

UNLABELLED: Orthoreovirus fusion-associated small transmembrane (FAST) proteins are dedicated cell-cell fusogens responsible for multinucleated syncytium formation and are virulence determinants of the fusogenic reoviruses. While numerous studies on the FAST proteins and enveloped-virus fusogens have delineated steps involved in membrane fusion and pore formation, little is known about the mechanics of pore expansion needed for syncytiogenesis. We now report that RNA interference (RNAi) knockdown of annexin A1 (AX1) expression dramatically reduced both reptilian reovirus p14 and measles virus F and H protein-mediated pore expansion during syncytiogenesis but had no effect on pore formation. A similar effect was obtained by chelating intracellular calcium, which dramatically decreased syncytiogenesis in the absence of detectable effects on p14-induced pore formation. Coimmunoprecipitation revealed calcium-dependent interaction between AX1 and p14 or measles virus F and H proteins, and fluorescence resonance energy transfer (FRET) demonstrated calcium-dependent p14-AX1 interactions in cellulo. Furthermore, antibody inhibition of extracellular AX1 had no effect on p14-induced syncytium formation but did impair cell-cell fusion mediated by the endogenous muscle cell fusion machinery in C2C12 mouse myoblasts. AX1 can therefore exert diverse, fusogen-specific effects on cell-cell fusion, functioning as an extracellular mediator of differentiation-dependent membrane fusion or as an intracellular promoter of postfusion pore expansion and syncytium formation following virus-mediated cell-cell fusion. IMPORTANCE: Numerous enveloped viruses and nonenveloped fusogenic orthoreoviruses encode membrane fusion proteins that induce syncytium formation, which has been linked to viral pathogenicity. Considerable insights into the mechanisms of membrane fusion have been obtained, but processes that drive postfusion expansion of fusion pores to generate syncytia are poorly understood. This study identifies intracellular calcium and annexin A1 (AX1) as key factors required for efficient pore expansion during syncytium formation mediated by the reptilian reovirus p14 and measles virus F and H fusion protein complexes. Involvement of intracellular AX1 in syncytiogenesis directly correlates with a requirement for intracellular calcium in p14-AX1 interactions and pore expansion but not membrane fusion and pore formation. This is the first demonstration that intracellular AX1 is involved in pore expansion, which suggests that the AX1 pathway may be a common host cell response needed to resolve virus-induced cell-cell fusion pores.


Assuntos
Anexina A1/metabolismo , Cálcio/metabolismo , Regulação Viral da Expressão Gênica/genética , Células Gigantes/virologia , Vírus do Sarampo/metabolismo , Orthoreovirus/metabolismo , Proteínas Virais/metabolismo , Animais , Fusão Celular , Linhagem Celular , Chlorocebus aethiops , DNA Complementar/genética , Fibroblastos , Transferência Ressonante de Energia de Fluorescência , Regulação Viral da Expressão Gênica/fisiologia , Células Gigantes/fisiologia , Proteínas de Fluorescência Verde , Humanos , Camundongos , Orthoreovirus/patogenicidade , Plasmídeos/genética , Codorniz , Interferência de RNA , Células Vero , Proteínas Virais de Fusão/metabolismo , Virulência
17.
Mol Biol Cell ; 25(6): 866-78, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24451258

RESUMO

Although numerous linear motifs that direct protein trafficking within cells have been identified, there are few examples of linear sorting signals mediating directed export of membrane proteins from the Golgi complex to the plasma membrane. The reovirus fusion-associated small transmembrane proteins are simple, single-pass transmembrane proteins that traffic through the endoplasmic reticulum-Golgi pathway to the plasma membrane, where they induce cell-cell membrane fusion. Here we show that a membrane-proximal, polybasic motif (PBM) in the cytosolic tail of p14 is essential for efficient export of p14 from the Golgi complex to the plasma membrane. Extensive mutagenic analysis reveals that the number, but not the identity or position, of basic residues present in the PBM dictates p14 export from the Golgi complex, with a minimum of three basic residues required for efficient Golgi export. Results further indicate that the tribasic motif does not affect plasma membrane retention of p14. Furthermore, introduction of the tribasic motif into a Golgi-localized, chimeric ERGIC-53 protein directs export from the Golgi complex to the plasma membrane. The p14 PBM is the first example of an autonomous, tribasic signal required for Golgi export to the plasma membrane.


Assuntos
Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Sinais Direcionadores de Proteínas/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Virais/metabolismo , Motivos de Aminoácidos , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Regulação da Expressão Gênica , Dados de Sequência Molecular , Transporte Proteico , Codorniz , Proteínas Recombinantes de Fusão/genética , Reoviridae/genética , Reoviridae/metabolismo , Transdução de Sinais , Células Vero , Proteínas Virais/genética
18.
FASEB J ; 27(12): 5046-58, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24005909

RESUMO

Myopodin is an actin-binding protein that promotes cancer cell migration in response to serum stimulation and is associated with invasive tumor development. To determine whether enhanced migration reflects changes in actin cytoskeleton remodeling, fluorescence confocal microscopy was used to examine the composition and morphology of filamentous actin structures in mock-transduced cells vs. stably transduced PC3 cells expressing human myopodin isoforms, and the chemokinetic response of cells was quantified using transwell assays. The same approaches were used to analyze the effects of external migration stimuli, actin polymerization inhibitors or deletion of the isoform-specific amino- and/or carboxy termini on cell migration and actin bundle formation. Results indicate that the termini of the myopodin isoforms differentially alter the formation of morphologically distinct F-actin networks that also differ in their myosin and myopodin staining patterns. Furthermore, enhanced cell migration was reduced by >50% when actin bundle formation was impaired by myopodin-truncation, low concentrations of an actin polymerization inhibitor, or in the absence of an external migration stimulus. Human myopodin isoforms are therefore potent regulators of stress fiber formation, inducing the formation of biochemically and morphologically distinct F-actin networks in the cell body whose presence directly correlates with increased cell migration.


Assuntos
Movimento Celular , Proteínas dos Microfilamentos/metabolismo , Fibras de Estresse/metabolismo , Linhagem Celular Tumoral , Humanos , Proteínas dos Microfilamentos/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Fibras de Estresse/ultraestrutura
19.
J Gen Virol ; 94(Pt 5): 1039-1050, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23343626

RESUMO

Piscine reovirus (PRV) is a tentative new member of the family Reoviridae and has been linked to heart and skeletal muscle inflammation in farmed Atlantic salmon (Salmo salar L.). Recent sequence-based evidence suggests that PRV is about equally related to members of the genera Orthoreovirus and Aquareovirus. Sequence similarities have also suggested that PRV might encode a fusion-associated small transmembrane (FAST) protein, which in turn suggests that PRV might be the prototype of a new genus with syncytium-inducing potential. In previous support of this designation has been the absence of identifiable PRV-encoded homologues of either the virion outer-clamp protein of ortho- and aquareoviruses or the virion outer-fibre protein of most orthoreoviruses. In the current report, we have provided experimental evidence that the putative p13 FAST protein of PRV lacks the defining feature of the FAST protein family - the ability to induce syncytium formation. Instead, p13 is the first example of a cytosolic, integral membrane protein encoded by ortho- or aquareoviruses, and induces cytotoxicity in the absence of cell-cell fusion. Sequence analysis also identified signature motifs of the outer-clamp and outer-fibre proteins of other reoviruses in two of the predicted PRV gene products. Based on these findings, we conclude that PRV does not encode a FAST protein and is therefore unlikely to be a new fusogenic reovirus. The presence of a novel integral membrane protein and two previously unrecognized, essential outer-capsid proteins has important implications for the biology, evolution and taxonomic classification of this virus.


Assuntos
Proteínas do Capsídeo/genética , Doenças dos Peixes/virologia , Proteínas de Membrana/genética , Infecções por Reoviridae/veterinária , Reoviridae/classificação , Salmão , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteínas do Capsídeo/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Citoplasma , Células Gigantes , Proteínas de Membrana/metabolismo , Dados de Sequência Molecular , Orthoreovirus/classificação , Orthoreovirus/genética , Orthoreovirus/isolamento & purificação , Orthoreovirus/metabolismo , Filogenia , Proteínas Recombinantes de Fusão , Reoviridae/química , Reoviridae/genética , Reoviridae/isolamento & purificação , Infecções por Reoviridae/virologia , Alinhamento de Sequência , Células Vero , Vírion
20.
Carcinogenesis ; 33(11): 2100-7, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22915763

RESUMO

The gene encoding myopodin, an actin binding protein, is commonly deleted in invasive, but not in indolent, prostate cancers. There are conflicting reports on the effects of myopodin expression on prostate cancer cell migration and invasion. The recent recognition that myopodin is expressed as four different isoforms further complicates our understanding of how this potentially important invasive prostate cancer biomarker affects tumor cell migration and invasion. We now show that myopodin affects the chemokinetic, rather than the chemotactic, properties of PC3 prostate cancer cells. Furthermore, all myopodin isoforms can either increase or decrease PC3 cell migration in response to different chemokinetic stimuli. These migration properties were reflected by differences in cell morphology and the relative dependence on Rho-ROCK signaling pathways induced by the environmental stimuli. Truncation analysis determined that a unique 9-residue C-terminal sequence in the shortest isoform and the conserved, PDZ domain-containing N-terminal region of the long isoforms both contribute to the ability of myopodin to alter the response of PC3 cells to chemokinetic stimuli. Matrigel invasion assays also indicated that myopodin primarily affects the migration, rather than the invasion, properties of PC3 cells. The correlation between loss of myopodin expression and invasive prostate cancer therefore reflects complex myopodin interactions with pathways that regulate the cellular migration response to diverse signals that may be present in a tumor microenvironment.


Assuntos
Movimento Celular/efeitos dos fármacos , Quimiocinas/farmacologia , Proteínas dos Microfilamentos/metabolismo , Neoplasias da Próstata/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Quinases Associadas a rho/metabolismo , Animais , Western Blotting , Movimento Celular/fisiologia , Células Cultivadas , Clonagem Molecular , Humanos , Imunoprecipitação , Masculino , Camundongos , Cadeias Pesadas de Miosina/metabolismo , Miosina Tipo I/metabolismo , Miosina Tipo III/metabolismo , Células NIH 3T3 , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Isoformas de Proteínas , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA