Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 13(1): 5813, 2023 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-37037845

RESUMO

Half of the marine virosphere is hypothesized to be RNA viruses (kingdom Orthornavirae) that infect abundant micro-eukaryotic hosts (e.g. protists). To test this, quantitative approaches that broadly track infections in situ are needed. Here, we describe a technique-dsRNA-Immunofluorescence (dsRIF)-that uses a double-stranded RNA (dsRNA) targeting monoclonal antibody to assess host infection status based on the presence of dsRNA, a replicative intermediate of all Orthornavirae infections. We show that the dinoflagellate Heterocapsa circularisquama produces dsRIF signal ~ 1000 times above background autofluorescence when infected by the + ssRNA virus HcRNAV. dsRNA-positive virocells were detected across > 50% of the 48-h infection cycle and accumulated to represent at least 63% of the population. Photosynthetic and chromosomal integrity remained intact during peak replication, indicating HcRNAV infection does not interrupt these processes. This work validates the use of dsRIF on marine RNA viruses and their hosts, setting the stage for quantitative environmental applications that will accelerate understanding of virus-driven ecosystem impacts.


Assuntos
Dinoflagellida , Infecções por Vírus de RNA , Vírus de RNA , Vírus , Humanos , RNA Viral/genética , Ecossistema , Vírus de RNA/genética , Vírus/genética , Dinoflagellida/genética , RNA de Cadeia Dupla
2.
J Biol Chem ; 299(3): 102912, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36649910

RESUMO

Daptomycin (DAP) is an antibiotic frequently used as a drug of last resort against vancomycin-resistant enterococci. One of the major challenges when using DAP against vancomycin-resistant enterococci is the emergence of resistance, which is mediated by the cell-envelope stress system LiaFSR. Indeed, inhibition of LiaFSR signaling has been suggested as a strategy to "resensitize" enterococci to DAP. In the absence of LiaFSR, alternative pathways mediating DAP resistance have been identified, including adaptive mutations in the enolpyruvate transferase MurAA (MurAAA149E), which catalyzes the first committed step in peptidoglycan biosynthesis; however, how these mutations confer resistance is unclear. Here, we investigated the biochemical basis for MurAAA149E-mediated adaptation to DAP to determine whether such an alternative pathway would undermine the potential efficacy of therapies that target the LiaFSR pathway. We found cells expressing MurAAA149E had increased susceptibility to glycoside hydrolases, consistent with decreased cell wall integrity. Furthermore, structure-function studies of MurAA and MurAAA149E using X-ray crystallography and biochemical analyses indicated only a modest decrease in MurAAA149E activity, but a 16-fold increase in affinity for MurG, which performs the last intracellular step of peptidoglycan synthesis. Exposure to DAP leads to mislocalization of cell division proteins including MurG. In Bacillus subtilis, MurAA and MurG colocalize at division septa and, thus, we propose MurAAA149E may contribute to DAP nonsusceptibility by increasing the stability of MurAA-MurG interactions to reduce DAP-induced mislocalization of these essential protein complexes.


Assuntos
Daptomicina , Enterococcus faecium , Transferases , Antibacterianos/farmacologia , Antibacterianos/metabolismo , Proteínas de Bactérias/metabolismo , Daptomicina/metabolismo , Daptomicina/farmacologia , Farmacorresistência Bacteriana , Enterococcus faecium/efeitos dos fármacos , Enterococcus faecium/metabolismo , Testes de Sensibilidade Microbiana , Peptidoglicano/metabolismo , Transferases/metabolismo
3.
Nat Commun ; 13(1): 3057, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35650195

RESUMO

Desmoplastic small round cell tumor (DSRCT) is an aggressive, usually incurable sarcoma subtype that predominantly occurs in post-pubertal young males. Recent evidence suggests that the androgen receptor (AR) can promote tumor progression in DSRCTs. However, the mechanism of AR-induced oncogenic stimulation remains undetermined. Herein, we demonstrate that enzalutamide and AR-directed antisense oligonucleotides (AR-ASO) block 5α-dihydrotestosterone (DHT)-induced DSRCT cell proliferation and reduce xenograft tumor burden. Gene expression analysis and chromatin immunoprecipitation sequencing (ChIP-seq) were performed to elucidate how AR signaling regulates cellular epigenetic programs. Remarkably, ChIP-seq revealed novel DSRCT-specific AR DNA binding sites adjacent to key oncogenic regulators, including WT1 (the C-terminal partner of the pathognomonic fusion protein) and FOXF1. Additionally, AR occupied enhancer sites that regulate the Wnt pathway, neural differentiation, and embryonic organ development, implicating AR in dysfunctional cell lineage commitment. Our findings have direct clinical implications given the widespread availability of FDA-approved androgen-targeted agents used for prostate cancer.


Assuntos
Antagonistas de Receptores de Andrógenos , Tumor Desmoplásico de Pequenas Células Redondas , Receptores Androgênicos , Antagonistas de Receptores de Andrógenos/farmacologia , Androgênios , Animais , Linhagem Celular Tumoral , Tumor Desmoplásico de Pequenas Células Redondas/genética , Humanos , Masculino , Oligonucleotídeos Antissenso/farmacologia , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Cancer Gene Ther ; 28(12): 1325-1338, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33408328

RESUMO

Osteosarcoma (OS) is a molecularly heterogeneous, aggressive, poorly differentiated pediatric bone cancer that frequently spreads to the lung. Relatively little is known about phenotypic and epigenetic changes that promote lung metastases. To identify key drivers of metastasis, we studied human CCH-OS-D OS cells within a previously described rat acellular lung (ACL) model that preserves the native lung architecture, extracellular matrix, and capillary network. This system identified a subset of cells-termed derived circulating tumor cells (dCTCs)-that can migrate, intravasate, and spread within a bioreactor-perfused capillary network. Remarkably, dCTCs highly expressed epithelial-to-mesenchymal transition (EMT)-associated transcription factors (EMT-TFs), such as ZEB1, TWIST, and SOX9, which suggests that they undergo cellular reprogramming toward a less differentiated state by coopting the same epigenetic machinery used by carcinomas. Since YAP/TAZ and AXL tightly regulate the fate and plasticity of normal mesenchymal cells in response to microenvironmental cues, we explored whether these proteins contributed to OS metastatic potential using an isogenic pair of human OS cell lines that differ in AXL expression. We show that AXL inhibition significantly reduced the number of MG63.2 pulmonary metastases in murine models. Collectively, we present a laboratory-based method to detect and characterize a pure population of dCTCs, which provides a unique opportunity to study how OS cell fate and differentiation contributes to metastatic potential. Though the important step of clinical validation remains, our identification of AXL, ZEB1, and TWIST upregulation raises the tantalizing prospect that EMT-TF-directed therapies might expand the arsenal of therapies used to combat advanced-stage OS.


Assuntos
Osteossarcoma/genética , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Proteínas de Sinalização YAP/metabolismo , Animais , Desdiferenciação Celular , Modelos Animais de Doenças , Humanos , Camundongos , Metástase Neoplásica , Osteossarcoma/patologia , Receptor Tirosina Quinase Axl
6.
Sci Rep ; 7: 44667, 2017 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-28317850

RESUMO

Mammalian parental genomes contribute differently to early embryonic development. Before activation of the zygotic genome, the maternal genome provides all transcripts and proteins required for the transition from a highly specialized oocyte to a pluripotent embryo. Depletion of these maternally-encoded transcripts frequently results in failure of preimplantation embryonic development, but their functions in this process are incompletely understood. We found that female mice lacking NLRP2 are subfertile because of early embryonic loss and the production of fewer offspring that have a wide array of developmental phenotypes and abnormal DNA methylation at imprinted loci. By demonstrating that NLRP2 is a member of the subcortical maternal complex (SCMC), an essential cytoplasmic complex in oocytes and preimplantation embryos with poorly understood function, we identified imprinted postzygotic DNA methylation maintenance, likely by directing subcellular localization of proteins involved in this process, such as DNMT1, as a new crucial role of the SCMC for mammalian reproduction.


Assuntos
Reprogramação Celular/genética , Desenvolvimento Embrionário , Epigênese Genética , Fertilidade , Complexos Multiproteicos/metabolismo , Proteínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas Reguladoras de Apoptose , Blastocisto/metabolismo , Forma Celular , Metilação de DNA/genética , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário/genética , Feminino , Fertilidade/genética , Fertilização/genética , Impressão Genômica , Masculino , Camundongos , Oócitos/citologia , Oócitos/metabolismo , Gravidez , Resultado da Gravidez , Ligação Proteica
7.
Methods ; 90: 28-38, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26344758

RESUMO

Calcium signaling is a ubiquitous and versatile process involved in nearly every cellular process, and exploitation of host calcium signals is a common strategy used by viruses to facilitate replication and cause disease. Small molecule fluorescent calcium dyes have been used by many to examine changes in host cell calcium signaling and calcium channel activation during virus infections, but disadvantages of these dyes, including poor loading and poor long-term retention, complicate analysis of calcium imaging in virus-infected cells due to changes in cell physiology and membrane integrity. The recent expansion of genetically-encoded calcium indicators (GECIs), including blue and red-shifted color variants and variants with calcium affinities appropriate for calcium storage organelles like the endoplasmic reticulum (ER), make the use of GECIs an attractive alternative for calcium imaging in the context of virus infections. Here we describe the development and testing of cell lines stably expressing both green cytoplasmic (GCaMP5G and GCaMP6s) and red ER-targeted (RCEPIAer) GECIs. Using three viruses (rotavirus, poliovirus and respiratory syncytial virus) previously shown to disrupt host calcium homeostasis, we show the GECI cell lines can be used to detect simultaneous cytoplasmic and ER calcium signals. Further, we demonstrate the GECI expression has sufficient stability to enable long-term confocal imaging of both cytoplasmic and ER calcium during the course of virus infections.


Assuntos
Sinalização do Cálcio , Cálcio/análise , Interações Hospedeiro-Patógeno , Microscopia de Fluorescência/métodos , Animais , Linhagem Celular , Chlorocebus aethiops , Retículo Endoplasmático/metabolismo , Células HEK293 , Humanos , Indicadores e Reagentes/química , Poliovirus/metabolismo , Vírus Sinciciais Respiratórios/metabolismo , Rotavirus/metabolismo
8.
J Virol ; 87(24): 13579-88, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24109210

RESUMO

Rotavirus nonstructural protein 4 (NSP4) induces dramatic changes in cellular calcium homeostasis. These include increased endoplasmic reticulum (ER) permeability, resulting in decreased ER calcium stores and activation of plasma membrane (PM) calcium influx channels, ultimately causing a 2- to 4-fold elevation in cytoplasmic calcium. Elevated cytoplasmic calcium is absolutely required for virus replication, but the underlying mechanisms responsible for calcium influx remain poorly understood. NSP4 is an ER-localized viroporin, whose activity depletes ER calcium, which ultimately leads to calcium influx. We hypothesized that NSP4-mediated depletion of ER calcium activates store-operated calcium entry (SOCE) through activation of the ER calcium sensor stromal interaction molecule 1 (STIM1). We established and used a stable yellow fluorescent protein-expressing STIM1 cell line (YFP-STIM1) as a biosensor to assess STIM1 activation (puncta formation) by rotavirus infection and NSP4 expression. We found that STIM1 is constitutively active in rotavirus-infected cells and that STIM1 puncta colocalize with the PM-localized Orai1 SOCE calcium channel. Expression of wild-type NSP4 activated STIM1, resulting in PM calcium influx, but an NSP4 viroporin mutant failed to induce STIM1 activation and did not activate the PM calcium entry pathway. Finally, knockdown of STIM1 significantly reduced rotavirus yield, indicating STIM1 plays a critical role in virus replication. These data demonstrate that while rotavirus may ultimately activate multiple calcium channels in the PM, calcium influx is predicated on NSP4 viroporin-mediated activation of STIM1 in the ER. This is the first report of viroporin-mediated activation of SOCE, reinforcing NSP4 as a robust model to understand dysregulation of calcium homeostasis during virus infections.


Assuntos
Cálcio/metabolismo , Retículo Endoplasmático/metabolismo , Glicoproteínas/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Infecções por Rotavirus/metabolismo , Infecções por Rotavirus/virologia , Rotavirus/metabolismo , Toxinas Biológicas/metabolismo , Proteínas não Estruturais Virais/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Citoplasma/metabolismo , Retículo Endoplasmático/genética , Glicoproteínas/genética , Humanos , Transporte de Íons , Proteínas de Membrana/genética , Proteínas de Neoplasias/genética , Rotavirus/genética , Infecções por Rotavirus/genética , Molécula 1 de Interação Estromal , Toxinas Biológicas/genética , Proteínas não Estruturais Virais/genética
9.
PLoS One ; 7(12): e50830, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23239987

RESUMO

BACKGROUND: Serum antibody responses in humans to inactivated influenza A (H5N1), (H9N2) and A (H7) vaccines have been varied but frequently low, particularly for subunit vaccines without adjuvant despite hemagglutinin (HA) concentrations expected to induce good responses. DESIGN: To help understand the low responses to subunit vaccines, we evaluated influenza A (H5N1), (H9N2), (H7N7) vaccines and 2009 pandemic (H1N1) vaccines for antigen uptake, processing and presentation by dendritic cells to T cells, conformation of vaccine HA in antibody binding assays and gel analyses, HA titers with different red blood cells, and vaccine morphology in electron micrographs (EM). RESULTS: Antigen uptake, processing and presentation of H5, H7, H9 and H1 vaccine preparations evaluated in humans appeared normal. No differences were detected in antibody interactions with vaccine and matched virus; although H7 trimer was not detected in western blots, no abnormalities in the conformation of the HA antigens were identified. The lowest HA titers for the vaccines were <1:4 for the H7 vaccine and 1:661 for an H9 vaccine; these vaccines induced the fewest antibody responses. A (H1N1) vaccines were the most immunogenic in humans; intact virus and virus pieces were prominent in EM. A good immunogenic A (H9N2) vaccine contained primarily particles of viral membrane with external HA and NA. A (H5N1) vaccines intermediate in immunogenicity were mostly indistinct structural units with stellates; the least immunogenic A (H7N7) vaccine contained mostly small 5 to 20 nm structures. SUMMARY: Antigen uptake, processing and presentation to human T cells and conformation of the HA appeared normal for each inactivated influenza A vaccine. Low HA titer was associated with low immunogenicity and presence of particles or split virus pieces was associated with higher immunogenicity.


Assuntos
Vacinas contra Influenza , Influenza Aviária , Influenza Humana , Vacinas de Produtos Inativados , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Apresentação de Antígeno/imunologia , Aves/imunologia , Aves/virologia , Testes de Inibição da Hemaglutinação , Humanos , Virus da Influenza A Subtipo H5N1/imunologia , Vírus da Influenza A Subtipo H7N7/imunologia , Vírus da Influenza A Subtipo H9N2/imunologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Influenza Aviária/imunologia , Influenza Aviária/prevenção & controle , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/imunologia
10.
Proc Natl Acad Sci U S A ; 109(50): E3405-13, 2012 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-23184977

RESUMO

Autophagy is a cellular degradation process involving an intracellular membrane trafficking pathway that recycles cellular components or eliminates intracellular microbes in lysosomes. Many pathogens subvert autophagy to enhance their replication, but the mechanisms these pathogens use to initiate the autophagy process have not been elucidated. This study identifies rotavirus as a pathogen that encodes a viroporin, nonstructural protein 4, which releases endoplasmic reticulum calcium into the cytoplasm, thereby activating a calcium/calmodulin-dependent kinase kinase-ß and 5' adenosine monophosphate-activated protein kinase-dependent signaling pathway to initiate autophagy. Rotavirus hijacks this membrane trafficking pathway to transport viral proteins from the endoplasmic reticulum to sites of viral replication to produce infectious virus. This process requires PI3K activity and autophagy-initiation proteins Atg3 and Atg5, and it is abrogated by chelating cytoplasmic calcium or inhibiting calcium/calmodulin-dependent kinase kinase-ß. Although the early stages of autophagy are initiated, rotavirus infection also blocks autophagy maturation. These studies identify a unique mechanism of virus-mediated, calcium-activated signaling that initiates autophagy and hijacks this membrane trafficking pathway to transport viral proteins to sites of viral assembly.


Assuntos
Autofagia/fisiologia , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Rotavirus/fisiologia , Replicação Viral/fisiologia , Animais , Proteína 5 Relacionada à Autofagia , Proteínas Relacionadas à Autofagia , Sinalização do Cálcio , Linhagem Celular , Células Cultivadas , Ativação Enzimática , Glicoproteínas/fisiologia , Macaca mulatta , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/deficiência , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/fisiologia , Transporte Proteico , Rotavirus/patogenicidade , Transdução de Sinais , Toxinas Biológicas/fisiologia , Enzimas de Conjugação de Ubiquitina/deficiência , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/fisiologia , Resposta a Proteínas não Dobradas , Proteínas não Estruturais Virais/fisiologia , Montagem de Vírus/fisiologia
11.
Virol J ; 9: 181, 2012 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-22943503

RESUMO

BACKGROUND: Our previous report that the Norwalk virus nonstructural protein p22 is an antagonist of the cellular secretory pathway suggests a new aspect of norovirus/host interaction. To explore conservation of function of this highly divergent calicivirus protein, we examined the effects of p22 homologues from four human and two murine noroviruses, and feline calicivirus on the secretory pathway. FINDINGS: All human noroviruses examined induced Golgi disruption and inhibited protein secretion, with the genogroup II.4 Houston virus being the most potent antagonist. Genogroup II.6 viruses have a conserved mutation in the mimic of an Endoplasmic Reticulum export signal (MERES) motif that is highly conserved in human norovirus homologues of p22 and is critical for secretory pathway antagonism, and these viruses had reduced levels of Golgi disruption and inhibition of protein secretion. p22 homologues from both persistent and nonpersistent strains of murine norovirus induced Golgi disruption, but only mildly inhibited cellular protein secretion. Feline calicivirus p30 did not induce Golgi disruption or inhibit cellular protein secretion. CONCLUSIONS: These differences confirm a norovirus-specific effect on host cell secretory pathway antagonism by homologues of p22, which may affect viral replication and/or cellular pathogenesis.


Assuntos
Interações Hospedeiro-Patógeno , Vírus Norwalk/genética , Vírus Norwalk/patogenicidade , Via Secretória , Proteínas não Estruturais Virais/metabolismo , Fatores de Virulência/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Complexo de Golgi/virologia , Humanos , Dados de Sequência Molecular , Alinhamento de Sequência , Proteínas não Estruturais Virais/genética , Fatores de Virulência/genética
12.
mBio ; 3(4): e00159-12, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22761392

RESUMO

UNLABELLED: Directed differentiation of stem cell lines into intestine-like tissue called induced human intestinal organoids (iHIOs) is now possible (J. R. Spence, C. N. Mayhew, S. A. Rankin, M. F. Kuhar, J. E. Vallance, K. Tolle, E. E. Hoskins, V. V. Kalinichenko, S. I. Wells, A. M. Zorn, N. F. Shroyer, and J. M. Wells, Nature 470:105-109, 2011). We tested iHIOs as a new model to cultivate and study fecal viruses. Protocols for infection of iHIOs with a laboratory strain of rotavirus, simian SA11, were developed. Proof-of-principle analyses showed that iHIOs support replication of a gastrointestinal virus, rotavirus, on the basis of detection of nonstructural viral proteins (nonstructural protein 4 [NSP4] and NSP2) by immunofluorescence, increased levels of viral RNA by quantitative reverse transcription-PCR (qRT-PCR), and production of infectious progeny virus. iHIOs were also shown to support replication of 12/13 clinical rotavirus isolates directly from stool samples. An unexpected finding was the detection of rotavirus infection not only in the epithelial cells but also in the mesenchymal cell population of the iHIOs. This work demonstrates that iHIOs offer a promising new model to study rotaviruses and other gastrointestinal viruses. IMPORTANCE: Gastrointestinal viral infections are a major cause of illness and death in children and adults. The ability to fully understand how viruses interact with human intestinal cells in order to cause disease has been hampered by insufficient methods for growing many gastrointestinal viruses in the laboratory. Induced human intestinal organoids (iHIOs) are a promising new model for generating intestine-like tissue. This is the first report of a study using iHIOs to cultivate any microorganism, in this case, an enteric virus. The evidence that both laboratory and clinical rotavirus isolates can replicate in iHIOs suggests that this model would be useful not only for studies of rotaviruses but also potentially of other infectious agents. Furthermore, detection of rotavirus proteins in unexpected cell types highlights the promise of this system to reveal new questions about pathogenesis that have not been previously recognized or investigated in other intestinal cell culture models.


Assuntos
Intestinos/virologia , Organoides/virologia , Infecções por Rotavirus/virologia , Rotavirus/fisiologia , Células-Tronco/virologia , Virologia/métodos , Adulto , Diferenciação Celular , Células Cultivadas , Feminino , Humanos , Intestinos/citologia , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Organoides/citologia , Rotavirus/genética , Células-Tronco/citologia
13.
J Virol ; 86(9): 4921-34, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22357281

RESUMO

Nonstructural protein 4 (NSP4) viroporin activity is critical for the replication and assembly of serogroup A rotavirus (RVA); however, the dramatic primary sequence divergence of NSP4s across serogroups raises the possibility that viroporin activity is not a common feature among RVs. We tested for NSP4 viroporin activity from divergent strains, including RVA (EC and Ty-1), RVB (IDIR), and RVC (Cowden). Canonical viroporin motifs were identified in RVA, RVB, and RVC NSP4s, but the arrangement of basic residues and the amphipathic α-helices was substantially different between serogroups. Using Escherichia coli and mammalian cell expression, we showed that each NSP4 tested had viroporin activity, but serogroup-specific viroporin phenotypes were identified. Only mammalian RVA and RVC NSP4s induced BL21-pLysS E. coli cell lysis, a classical viroporin activity assay. In contrast, RVA, RVB, and RVC NSP4 expression was universally cytotoxic to E. coli and disrupted reduction-oxidation activities, as measured by a new redox dye assay. In mammalian cells, RVB and RVC NSP4s were initially localized in the endoplasmic reticulum (ER) and trafficked into punctate structures that were mutually exclusive with RVA NSP4. The punctate structures partially localized to the ER-Golgi intermediate compartment (ERGIC) but primarily colocalized with punctate LC3, a marker for autophagosomes. Similar to RVA NSP4, expression of RVB and RVC NSP4s significantly elevated cytosolic calcium levels, demonstrating that despite strong primary sequence divergence, RV NSP4 has maintained viroporin activity across serogroups A to C. These data suggest that elevated cytosolic calcium is a common critical process for all rotavirus strains.


Assuntos
Glicoproteínas/genética , Glicoproteínas/metabolismo , Rotavirus/genética , Rotavirus/metabolismo , Toxinas Biológicas/genética , Toxinas Biológicas/metabolismo , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Sequência de Aminoácidos , Animais , Cálcio/metabolismo , Chlorocebus aethiops , Escherichia coli/metabolismo , Escherichia coli/virologia , Glicoproteínas/imunologia , Espaço Intracelular/metabolismo , Dados de Sequência Molecular , Oxirredução , Estrutura Terciária de Proteína , Transporte Proteico , Rotavirus/classificação , Homologia de Sequência de Aminoácidos , Toxinas Biológicas/imunologia , Proteínas não Estruturais Virais/imunologia
14.
Mol Biol Cell ; 22(16): 2810-22, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21697506

RESUMO

The Golgi complex has been implicated as a possible component of endoplasmic reticulum (ER) glycoprotein quality control, although the elucidation of its exact role is lacking. ERManI, a putative ER resident mannosidase, plays a rate-limiting role in generating a signal that targets misfolded N-linked glycoproteins for ER-associated degradation (ERAD). Herein we demonstrate that the endogenous human homologue predominantly resides in the Golgi complex, where it is subjected to O-glycosylation. To distinguish the intracellular site where the glycoprotein ERAD signal is generated, a COPI-binding motif was appended to the N terminus of the recombinant protein to facilitate its retrograde translocation back to the ER. Partial redistribution of the modified ERManI was observed along with an accelerated rate at which N-linked glycans of misfolded α1-antitrypsin variant NHK were trimmed. Despite these observations, the rate of NHK degradation was not accelerated, implicating the Golgi complex as the site for glycoprotein ERAD substrate tagging. Taken together, these data provide a potential mechanistic explanation for the spatial separation by which glycoprotein quality control components operate in mammalian cells.


Assuntos
Glicoproteínas/metabolismo , Complexo de Golgi/metabolismo , Manosidases/metabolismo , Transporte Proteico , Proteólise , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais Murinos/química , Sítios de Ligação , Linhagem Celular , Cricetinae , Cricetulus , Retículo Endoplasmático/metabolismo , Glicosilação , Humanos , Manosidases/química , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Dobramento de Proteína , Estrutura Terciária de Proteína , Solubilidade
15.
mBio ; 1(5)2010 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-21151776

RESUMO

Many viruses alter intracellular calcium homeostasis. The rotavirus nonstructural protein 4 (NSP4), an endoplasmic reticulum (ER) transmembrane glycoprotein, increases intracellular levels of cytoplasmic Ca(2+) ([Ca(2+)]cyto) through a phospholipase C-independent pathway, which is required for virus replication and morphogenesis. However, the NSP4 domain and mechanism that increases [Ca(2+)]cyto are unknown. We identified an NSP4 domain (amino acids [aa] 47 to 90) that inserts into membranes and has structural characteristics of viroporins, a class of small hydrophobic viral proteins that disrupt membrane integrity and ion homeostasis to facilitate virus entry, assembly, or release. Mutational analysis showed that NSP4 viroporin activity was mediated by an amphipathic α-helical domain downstream of a conserved lysine cluster. The lysine cluster directed integral membrane insertion of the viroporin domain and was critical for viroporin activity. In epithelial cells, expression of wild-type NSP4 increased the levels of free cytoplasmic Ca(2+) by 3.7-fold, but NSP4 viroporin mutants maintained low levels of [Ca(2+)]cyto, were retained in the ER, and failed to form cytoplasmic vesicular structures, called puncta, which surround viral replication and assembly sites in rotavirus-infected cells. When [Ca(2+)]cyto was increased pharmacologically with thapsigargin, viroporin mutants formed puncta, showing that elevation of calcium levels and puncta formation are distinct functions of NSP4 and indicating that NSP4 directly or indirectly responds to elevated cytoplasmic calcium levels. NSP4 viroporin activity establishes the mechanism for NSP4-mediated elevation of [Ca(2+)]cyto, a critical event that regulates rotavirus replication and virion assembly.


Assuntos
Cálcio/metabolismo , Glicoproteínas/metabolismo , Porinas/metabolismo , Rotavirus/patogenicidade , Toxinas Biológicas/metabolismo , Proteínas não Estruturais Virais/metabolismo , Fatores de Virulência/metabolismo , Animais , Linhagem Celular , Citoplasma/química , Análise Mutacional de DNA , Retículo Endoplasmático/química , Homeostase , Humanos , Modelos Biológicos , Modelos Químicos , Estrutura Terciária de Proteína
16.
Proc Natl Acad Sci U S A ; 105(26): 8811-8, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18587047

RESUMO

Rotavirus NSP4 is a viral enterotoxin capable of causing diarrhea in neonatal mice. This process is initiated by the binding of extracellular NSP4 to target molecule(s) on the cell surface that triggers a signaling cascade leading to diarrhea. We now report that the integrins alpha1beta1 and alpha2beta1 are receptors for NSP4. NSP4 specifically binds to the alpha1 and alpha2 I domains with apparent K(d) = 1-2.7 muM. Binding is mediated by the I domain metal ion-dependent adhesion site motif, requires Mg(2+) or Mn(2+), is abolished with EDTA, and an NSP4 point mutant, E(120)A, fails to bind alpha2 integrin I domain. NSP4 has two distinct integrin interaction domains. NSP4 amino acids 114-130 are essential for binding to the I domain, and NSP4 peptide 114-135 blocks binding of the natural ligand, collagen I, to integrin alpha2. NSP4 amino acids 131-140 are not associated with the initial binding to the I domain, but elicit signaling that leads to the spreading of attached C2C12-alpha2 cells, mouse myoblast cells stably expressing the human alpha2 integrin. NSP4 colocalizes with integrin alpha2 on the basolateral surface of rotavirus-infected polarized intestinal epithelial (Caco-2) cells as well as surrounding noninfected cells. NSP4 mutants that fail to bind or signal through integrin alpha2 were attenuated in diarrhea induction in neonatal mice. These results indicate that NSP4 interaction with integrin alpha1 and alpha2 is an important component of enterotoxin function and rotavirus pathogenesis, further distinguishing this viral virulence factor from other microbial enterotoxins.


Assuntos
Enterotoxinas/metabolismo , Glicoproteínas/metabolismo , Integrina alfa1beta1/metabolismo , Integrina alfa2beta1/metabolismo , Rotavirus/metabolismo , Toxinas Biológicas/metabolismo , Proteínas não Estruturais Virais/metabolismo , Androstadienos/farmacologia , Animais , Sítios de Ligação , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Diarreia/induzido quimicamente , Diarreia/metabolismo , Enterotoxinas/química , Ensaio de Imunoadsorção Enzimática , Estrenos/farmacologia , Glicoproteínas/química , Humanos , Integrina alfa1beta1/química , Integrina alfa2beta1/química , Camundongos , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Transporte Proteico/efeitos dos fármacos , Pirrolidinonas/farmacologia , Rotavirus/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Ressonância de Plasmônio de Superfície , Toxinas Biológicas/química , Proteínas não Estruturais Virais/química , Wortmanina
17.
Circulation ; 114(1 Suppl): I200-5, 2006 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-16820572

RESUMO

BACKGROUND: Thoracic aortic dissection (TAD) is characterized by dysregulated extracellular matrix. Little is known about the alterations of collagen and stimulators of collagen synthesis, eg, connective tissue growth factor (CTGF), in patients with TAD. In this study, we examined their roles in TAD. METHODS AND RESULTS: Surgical specimens of the aortic wall of TAD patients (n=10) and controls (n=10) were tested for collagen types I and III and CTGF expression. When compared with controls, protein levels of type I and III collagen and CTGF were significantly increased by 3.2-, 3.7-, and 5.3-fold, respectively (P<0.05 for all). Similar patterns were shown in mRNA levels of type Ialpha and Ialpha2 collagen and CTGF. Using immunohistochemistry and trichrome staining, we also observed elevated levels of collagen in the aortic media and adventitia. Treatment with recombinant human CTGF increased collagen synthesis in cultured aortic smooth muscle cells in a dose- and time-dependent fashion, in which expression of collagens increased from 506+/-108 counts per minute to 2764+/-240 cpm by 50 ng/mL CTGF, and from 30+/-43 cpm to 429+/-102 cpm at 48 hours. CONCLUSIONS: TAD patients exhibited significantly increased expression of aortic collagen types I and III as well as CTGF, which is likely to be responsible for the compromised aortic distensibility and systemic compliance. Because CTGF can increase collagen expression, CTGF may be a new target molecule in the pathogenesis and progression of TAD.


Assuntos
Aneurisma da Aorta Torácica/metabolismo , Dissecção Aórtica/metabolismo , Colágeno Tipo III/biossíntese , Colágeno Tipo I/biossíntese , Proteínas Imediatamente Precoces/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Adulto , Idoso , Dissecção Aórtica/genética , Dissecção Aórtica/patologia , Aorta Torácica/metabolismo , Aorta Torácica/patologia , Aneurisma da Aorta Torácica/genética , Aneurisma da Aorta Torácica/patologia , Apoptose , Colágeno Tipo I/genética , Colágeno Tipo III/genética , Complacência (Medida de Distensibilidade) , Fator de Crescimento do Tecido Conjuntivo , Feminino , Perfilação da Expressão Gênica , Humanos , Proteínas Imediatamente Precoces/biossíntese , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Masculino , Pessoa de Meia-Idade , Músculo Liso Vascular/patologia , Proteínas Recombinantes/farmacologia
18.
FEBS Lett ; 580(11): 2779-87, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16650413

RESUMO

Human cytomegalovirus (HCMV) exerts anti-apoptotic effect during early stage of infection, which provides HCMV time for propagation. We investigated pathways mediating the resistance to H(2)O(2)-induced cell death - a self-defense mechanism to remove infected cells. We found that human aortic endothelial cells (HAECs) infected with VHL/E strain of HCMV during first 3 days were resistant to H(2)O(2) (0-2 mM) induced apoptosis. This anti-apoptotic effect may be mediated by the upregulation of Bcl-2, an anti-apoptotic protein through the activation pro-survival pathway extracellular signal regulated kinase (ERK). Through this mechanism, HCMV is able to propagate and causes endothelial dysfunction, hence vascular disease.


Assuntos
Apoptose/efeitos dos fármacos , Citomegalovirus/fisiologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/virologia , Peróxido de Hidrogênio/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Caspase 3 , Caspases/metabolismo , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/enzimologia , Ativação Enzimática/efeitos dos fármacos , Humanos , Membranas Intracelulares/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/genética , Regulação para Cima
19.
J Cell Sci ; 119(Pt 12): 2457-67, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16720642

RESUMO

Human cytomegalovirus (HCMV) infection results in endothelial dysfunction, typically known as dysregulated apoptosis, and aberrant expression and sub-cellular localization of p53, a tumor suppressor that accumulates at the late stage of infection. In this study, we examined three hypotheses that could be responsible for HCMV-induced cytoplasmic p53 accumulation at the later stage of infection: hyperactive nuclear export, cytoplasmic p53 tethering and delayed p53 degradation. Leptomycin B treatment, a nuclear export inhibitor, was unable to reduce cytoplasmic p53, thereby eliminating the hyperactive nuclear export mechanism. The findings that nascent p53 still entered nuclei after the nuclear export inhibition indicated that cytoplasmic tethering may play a minor role. Cytoplasmic p53 was still observed after the translation activities were blocked by cycloheximide. There was more than an eight-fold increase in the cytoplasmic p53 half-life with abnormal p53 ubiquitination. Taken together, these results suggest that delayed degradation could be responsible for the cytoplasmic p53 accumulation. The general slow-down of the proteasomal activity and the dysregulated p53 ubiquitination process at the later stage of infection could contribute to the reduced cytoplasmic p53 degradation and might be relevant to dysregulated endothelial apoptosis. The HCMV-induced changes in p53 dynamics could contribute to endothelial dysfunction.


Assuntos
Infecções por Citomegalovirus/virologia , Citomegalovirus/fisiologia , Citoplasma/metabolismo , Células Endoteliais/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Núcleo Celular/metabolismo , Núcleo Celular/virologia , Células Cultivadas , Cicloeximida/farmacologia , Citoplasma/virologia , Células Endoteliais/virologia , Humanos , Carioferinas/biossíntese , Proteínas Proto-Oncogênicas c-mdm2/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/biossíntese , Fatores de Tempo , Proteína Supressora de Tumor p53/efeitos dos fármacos , Ubiquitina/metabolismo , Proteína Exportina 1
20.
Cardiovasc Res ; 69(2): 502-11, 2006 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16316638

RESUMO

OBJECTIVES: Atherosclerosis is the leading cause of death in the United States, and human cytomegalovirus (HCMV) infection may play a role in the development of this disease. Diminished expression and/or activity of endothelial nitric oxide synthase (eNOS) are an early event in atherogenesis. In the current study, we investigated the effects of HCMV infection on eNOS activation in human aortic endothelial cells (HAECs). METHODS AND RESULTS: We found that HCMV inhibited eNOS phosphorylation/activation in HAECs. The signaling upstream of eNOS involving Akt and PDK1 were also suppressed by the HCMV infection. Moreover, HCMV infection increased the expression of PTEN (phosphatase and tensin homolog deleted on chromosome 10). Silencing PTEN expression with specific siRNA reversed the inhibitory effects on eNOS activation in HCMV-infected cells indicating the involvement of PTEN in mediating HCMV's inhibitory effects. Next we observed that the activation of p38 MAPK stress signaling pathway mediates HCMV's effects on PTEN up-regulation and eNOS inactivation. CONCLUSIONS: In summary, our findings suggest that inhibition of eNOS leading to endothelial dysfunction may be a basis of the pro-atherogenic effects of HCMV. Importantly, upregulation of PTEN and activation of stress signal p38 MAPK are involved in HCMV's inhibitory effects on eNOS activation.


Assuntos
Infecções por Citomegalovirus/metabolismo , Citomegalovirus , Endotélio Vascular/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Regulação para Cima , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Aterosclerose/metabolismo , Western Blotting/métodos , Células Cultivadas , Células Endoteliais , Ativação Enzimática , Imunofluorescência , Humanos , Sistema de Sinalização das MAP Quinases , Proteína Oncogênica v-akt/metabolismo , PTEN Fosfo-Hidrolase/genética , Proteínas Serina-Treonina Quinases/metabolismo , RNA Interferente Pequeno/genética , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA