Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 299(3): 102912, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36649910

RESUMEN

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.


Asunto(s)
Daptomicina , Enterococcus faecium , Transferasas , Antibacterianos/farmacología , Antibacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Daptomicina/metabolismo , Daptomicina/farmacología , Farmacorresistencia Bacteriana , Enterococcus faecium/efectos de los fármacos , Enterococcus faecium/metabolismo , Pruebas de Sensibilidad Microbiana , Peptidoglicano/metabolismo , Transferasas/metabolismo
2.
Methods ; 90: 28-38, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26344758

RESUMEN

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.


Asunto(s)
Señalización del Calcio , Calcio/análisis , Interacciones Huésped-Patógeno , Microscopía Fluorescente/métodos , Animales , Línea Celular , Chlorocebus aethiops , Retículo Endoplásmico/metabolismo , Células HEK293 , Humanos , Indicadores y Reactivos/química , Poliovirus/metabolismo , Virus Sincitiales Respiratorios/metabolismo , Rotavirus/metabolismo
3.
Proc Natl Acad Sci U S A ; 109(50): E3405-13, 2012 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-23184977

RESUMEN

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.


Asunto(s)
Autofagia/fisiología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Rotavirus/fisiología , Replicación Viral/fisiología , Animales , Proteína 5 Relacionada con la Autofagia , Proteínas Relacionadas con la Autofagia , Señalización del Calcio , Línea Celular , Células Cultivadas , Activación Enzimática , Glicoproteínas/fisiología , Macaca mulatta , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/fisiología , Transporte de Proteínas , Rotavirus/patogenicidad , Transducción de Señal , Toxinas Biológicas/fisiología , Enzimas Ubiquitina-Conjugadoras/deficiencia , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/fisiología , Respuesta de Proteína Desplegada , Proteínas no Estructurales Virales/fisiología , Ensamble de Virus/fisiología
4.
J Virol ; 87(24): 13579-88, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24109210

RESUMEN

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.


Asunto(s)
Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Glicoproteínas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Infecciones por Rotavirus/metabolismo , Infecciones por Rotavirus/virología , Rotavirus/metabolismo , Toxinas Biológicas/metabolismo , Proteínas no Estructurales Virales/metabolismo , Línea Celular , Membrana Celular/metabolismo , Citoplasma/metabolismo , Retículo Endoplásmico/genética , Glicoproteínas/genética , Humanos , Transporte Iónico , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética , Rotavirus/genética , Infecciones por Rotavirus/genética , Molécula de Interacción Estromal 1 , Toxinas Biológicas/genética , Proteínas no Estructurales Virales/genética
5.
J Virol ; 86(9): 4921-34, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22357281

RESUMEN

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.


Asunto(s)
Glicoproteínas/genética , Glicoproteínas/metabolismo , Rotavirus/genética , Rotavirus/metabolismo , Toxinas Biológicas/genética , Toxinas Biológicas/metabolismo , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Chlorocebus aethiops , Escherichia coli/metabolismo , Escherichia coli/virología , Glicoproteínas/inmunología , Espacio Intracelular/metabolismo , Datos de Secuencia Molecular , Oxidación-Reducción , Estructura Terciaria de Proteína , Transporte de Proteínas , Rotavirus/clasificación , Homología de Secuencia de Aminoácido , Toxinas Biológicas/inmunología , Proteínas no Estructurales Virales/inmunología
6.
Sci Rep ; 13(1): 5813, 2023 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-37037845

RESUMEN

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.


Asunto(s)
Dinoflagelados , Infecciones por Virus ARN , Virus ARN , Virus , Humanos , ARN Viral/genética , Ecosistema , Virus ARN/genética , Virus/genética , Dinoflagelados/genética , ARN Bicatenario
7.
Virol J ; 9: 181, 2012 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-22943503

RESUMEN

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.


Asunto(s)
Interacciones Huésped-Patógeno , Virus Norwalk/genética , Virus Norwalk/patogenicidad , Vías Secretoras , Proteínas no Estructurales Virales/metabolismo , Factores de Virulencia/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Aparato de Golgi/virología , Humanos , Datos de Secuencia Molecular , Alineación de Secuencia , Proteínas no Estructurales Virales/genética , Factores de Virulencia/genética
8.
Nat Commun ; 13(1): 3057, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35650195

RESUMEN

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.


Asunto(s)
Antagonistas de Receptores Androgénicos , Tumor Desmoplásico de Células Pequeñas Redondas , Receptores Androgénicos , Antagonistas de Receptores Androgénicos/farmacología , Andrógenos , Animales , Línea Celular Tumoral , Tumor Desmoplásico de Células Pequeñas Redondas/genética , Humanos , Masculino , Oligonucleótidos Antisentido/farmacología , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Proc Natl Acad Sci U S A ; 105(26): 8811-8, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18587047

RESUMEN

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.


Asunto(s)
Enterotoxinas/metabolismo , Glicoproteínas/metabolismo , Integrina alfa1beta1/metabolismo , Integrina alfa2beta1/metabolismo , Rotavirus/metabolismo , Toxinas Biológicas/metabolismo , Proteínas no Estructurales Virales/metabolismo , Androstadienos/farmacología , Animales , Sitios de Unión , Adhesión Celular/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Diarrea/inducido químicamente , Diarrea/metabolismo , Enterotoxinas/química , Ensayo de Inmunoadsorción Enzimática , Estrenos/farmacología , Glicoproteínas/química , Humanos , Integrina alfa1beta1/química , Integrina alfa2beta1/química , Ratones , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Transporte de Proteínas/efectos de los fármacos , Pirrolidinonas/farmacología , Rotavirus/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Resonancia por Plasmón de Superficie , Toxinas Biológicas/química , Proteínas no Estructurales Virales/química , Wortmanina
10.
Cancer Gene Ther ; 28(12): 1325-1338, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33408328

RESUMEN

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.


Asunto(s)
Osteosarcoma/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Señalizadoras YAP/metabolismo , Animales , Desdiferenciación Celular , Modelos Animales de Enfermedad , Humanos , Ratones , Metástasis de la Neoplasia , Osteosarcoma/patología , Tirosina Quinasa del Receptor Axl
11.
Circulation ; 114(1 Suppl): I200-5, 2006 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-16820572

RESUMEN

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.


Asunto(s)
Aneurisma de la Aorta Torácica/metabolismo , Disección Aórtica/metabolismo , Colágeno Tipo III/biosíntesis , Colágeno Tipo I/biosíntesis , Proteínas Inmediatas-Precoces/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Adulto , Anciano , Disección Aórtica/genética , Disección Aórtica/patología , Aorta Torácica/metabolismo , Aorta Torácica/patología , Aneurisma de la Aorta Torácica/genética , Aneurisma de la Aorta Torácica/patología , Apoptosis , Colágeno Tipo I/genética , Colágeno Tipo III/genética , Adaptabilidad , Factor de Crecimiento del Tejido Conjuntivo , Femenino , Perfilación de la Expresión Génica , Humanos , Proteínas Inmediatas-Precoces/biosíntesis , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/farmacología , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/farmacología , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/patología , Proteínas Recombinantes/farmacología
12.
Cardiovasc Res ; 69(2): 502-11, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16316638

RESUMEN

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.


Asunto(s)
Infecciones por Citomegalovirus/metabolismo , Citomegalovirus , Endotelio Vascular/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosfohidrolasa PTEN/metabolismo , Regulación hacia Arriba , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Aterosclerosis/metabolismo , Western Blotting/métodos , Células Cultivadas , Células Endoteliales , Activación Enzimática , Técnica del Anticuerpo Fluorescente , Humanos , Sistema de Señalización de MAP Quinasas , Proteína Oncogénica v-akt/metabolismo , Fosfohidrolasa PTEN/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño/genética , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
Sci Rep ; 7: 44667, 2017 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-28317850

RESUMEN

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.


Asunto(s)
Reprogramación Celular/genética , Desarrollo Embrionario , Epigénesis Genética , Fertilidad , Complejos Multiproteicos/metabolismo , Proteínas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Reguladoras de la Apoptosis , Blastocisto/metabolismo , Forma de la Célula , Metilación de ADN/genética , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/genética , Femenino , Fertilidad/genética , Fertilización/genética , Impresión Genómica , Masculino , Ratones , Oocitos/citología , Oocitos/metabolismo , Embarazo , Resultado del Embarazo , Unión Proteica
14.
FEBS Lett ; 580(11): 2779-87, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16650413

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Citomegalovirus/fisiología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/virología , Peróxido de Hidrógeno/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Caspasa 3 , Caspasas/metabolismo , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/enzimología , Activación Enzimática/efectos de los fármacos , Humanos , Membranas Intracelulares/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/genética , Regulación hacia Arriba
15.
Arterioscler Thromb Vasc Biol ; 25(12): 2509-14, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16210567

RESUMEN

BACKGROUND: Previously, we showed that the 27nt repeat polymorphism in endothelial nitric oxide synthase (eNOS) intron 4 was associated with altered eNOS mRNA and protein levels, nitric oxide (NO) production and vascular disease risk; the 27-nt repeats had a cis-acting role in eNOS promoter function. In the present study, we investigated nuclear protein that binds the 27nt repeat and mediates eNOS expression. METHODS AND RESULTS: Using 5'-biotin-labeled 27nt DNA duplex and streptavidin-agarose beads pull-down assay and mass spectrometry, we identified that nuclear beta-actin was one of the major 27nt binding proteins. Using the pGL3 reporter vectors containing the 5x27nt repeats as an enhancer in an in vitro transcription assay, we found that exogenous beta-actin significantly increased reporter gene transcription efficiency. The beta-actin's upregulating effect was compromised when exogenous 27nt RNA duplex was added. Furthermore, the eNOS expression was reduced when beta-actin gene was silenced by specific siRNA, and actin overexpression upregulated eNOS expression >3-fold. CONCLUSIONS: Our data demonstrate that beta-actin as a transcription factor stimulates eNOS expression; and the transcriptional effect appears to be 27nt-dependent. Our findings represent a novel molecular mechanism regulating eNOS expression, which could potentially lead to discoveries of eNOS specific pharmaceutical agents, eg, active peptides, with clinical applications.


Asunto(s)
Actinas/metabolismo , Endotelio Vascular/enzimología , Regulación Enzimológica de la Expresión Génica/fisiología , Óxido Nítrico Sintasa de Tipo III/genética , Factores de Transcripción/metabolismo , Actinas/genética , Aorta/citología , Núcleo Celular/fisiología , Células Cultivadas , Endotelio Vascular/citología , Silenciador del Gen , Genes Reporteros , Humanos , Intrones , Polimorfismo Genético , ARN Mensajero/metabolismo , Factores de Transcripción/genética , Transcripción Genética/fisiología
16.
FEBS Lett ; 579(3): 733-40, 2005 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-15670837

RESUMEN

Cigarette-induced endothelial dysfunction could be an early mediator of atherosclerosis. In this study, we explored the mechanisms of cigarette smoke extract (CSE)-induced human aortic endothelial cells (HAEC) apoptosis. We found that 10-65% of HAECs underwent apoptotic changes when HAECs were exposed to 0.001-0.02 cigarette equivalent unit of CSE for 4 h. CSE activated the caspases-3 and 8, the p38 MAP kinase and stress activated protein kinase/c-Jun N-terminal protein kinase (SAPK/JNK). Specific inhibitors of p38 MAP or SAPK/JNK reduced CSE-induced caspase activation. We further showed that eNOS pre-activation by L-arginine reduced endothelial apoptosis from 65% to 5%; and eNOS inhibition by N-omega-nitro-L-arginine methyl ester accentuated CSE-induced endothelial apoptosis. We suggest that appropriate endogenous NO production may be an important protective mechanism against smoking-induced endothelial damage.


Asunto(s)
Apoptosis , Endotelio Vascular/citología , Nicotiana , Óxido Nítrico/fisiología , Fumar , Secuencia de Bases , Western Blotting , Caspasa 3 , Caspasa 8 , Caspasas/metabolismo , Células Cultivadas , Cartilla de ADN , Endotelio Vascular/enzimología , Endotelio Vascular/metabolismo , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo III
17.
DNA Cell Biol ; 24(4): 218-24, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15812238

RESUMEN

DNA polymorphisms in endothelial nitric oxide synthase (eNOS) gene have been shown to be associated with constitutive eNOS expression and coronary artery disease (CAD). In the present study we explored the hypothesis whether genotype-dependent effects can be maintained in vitro during replication, or the effect is conditional on in vivo biological environments. Human umbilical vein endothelial cells (HUVEC) were collected and cultured from 89 normal deliveries of Mexican Americans. The cells were treated with or without cigarette smoking extracts (CSE) and genotypes of eNOS polymorphisms were determined by PCR. We measured the levels of eNOS by ELISA and its binding proteins including heat-shock protein 90 (Hsp-90) and caveolin-1 by Western blotting. The rare C allele for the promoter T786C polymorphism (0.2), and the rare 4 x 27-bp repeat allele in the intron 4 (0.30) were different from those reported in other populations. Yet, the rare T allele in the exon 7 (G894T polymorphism) was similar as others. After four passages in vitro, both the intron 4 and promoter polymorphisms maintained significant effects on eNOS mRNA levels in HUVECs (P < 0.05). However, the effects on eNOS protein and enzyme activity were less consistent. Although primary smokers had significantly lower eNOS protein levels (P < 0.05), the in vitro CSE treatment on cultured HUVECs only resulted in a significant reduction in NO levels as measured by the stable metabolites of nitrite/nitrate (P < 0.001). Neither Hsp-90 nor caveolin-1--important eNOS regulators--appears to mediate the genotypesmoking effects on eNOS expression although HUVECs did produce more Hsp-90 when exposed to CSE. Our study demonstrates that endothelial cells maintain genotype-dependent expression even after the deprivation of in vivo environment. However, the cigarette smoking-genotype interaction may require such in vivo conditions to be manifested.


Asunto(s)
Células Endoteliales/metabolismo , Expresión Génica/genética , Óxido Nítrico Sintasa/metabolismo , Polimorfismo Genético , ARN Mensajero/metabolismo , Venas Umbilicales/citología , Análisis de Varianza , Western Blotting , Caveolina 1 , Caveolinas/metabolismo , Células Cultivadas , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Expresión Génica/efectos de los fármacos , Genotipo , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Americanos Mexicanos , Mutación/genética , Nitratos/metabolismo , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo III , Nitritos/metabolismo , Reacción en Cadena de la Polimerasa , Breas/toxicidad
18.
Surgery ; 138(2): 352-9, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16153447

RESUMEN

BACKGROUND: Thoracic aortic aneurysmal diseases are characterized by degeneration of elastin within the aortic wall. Although proteinases, such as matrix metalloproteinase, appear to contribute to elastin degradation, little is known about the role of elastic fiber assembly in such diseases. Fibulin-5 is an extracellular protein that is expressed in the vascular basement membrane and regulates elastic fiber assembly by microfibril machinery. In this study, we examined whether thoracic aortic dissection (TAD) is associated with abnormal fibulin-5 expression. METHODS: Intraoperative aortic samples were obtained from 21 patients with proximal aortic dissection. Control aortic tissue was obtained from 11 organ donors, heart transplant recipients, and patients undergoing coronary artery bypass. An in vitro culture of vascular smooth muscle cells was obtained from 2 TAD patients and 1 control subject. To evaluate elastin expression, we stained tissue sections with Verhoeff-Van Gieson stain. Fibulin-5 messenger RNA (mRNA) expression was determined by quantitative real-time reverse-transcriptase-polymerase chain reaction. RESULTS: Aortic fibulin-5 mRNA and elastin content were decreased in TAD patients, compared with controls (P=.001 and P=.02, respectively). Decreased fibulin-5 expression strongly correlated with decreased amounts and fragmentation of elastin in aortic samples from patients with TAD (r=0.83, P < .0001 and F=20.7, P < .0001 respectively). The fibulin-5 mRNA in aortic vascular smooth muscle cells collected from TAD demonstrated a 38% decrease in expression, compared with the control. CONCLUSIONS: Patients with proximal aortic dissection exhibited significantly decreased expression of aortic fibulin-5. Decreased fibulin-5 may contribute to the pathogenesis of aortic dissection by impairing elastic fiber assembly.


Asunto(s)
Aneurisma de la Aorta Torácica/fisiopatología , Disección Aórtica/fisiopatología , Elastina/genética , Proteínas de la Matriz Extracelular/genética , Proteínas Recombinantes/genética , Anciano , Disección Aórtica/patología , Aorta Torácica/patología , Aorta Torácica/fisiología , Aneurisma de la Aorta Torácica/patología , Elastina/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , ARN Mensajero/análisis , Proteínas Recombinantes/metabolismo
19.
PLoS One ; 7(12): e50830, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23239987

RESUMEN

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.


Asunto(s)
Vacunas contra la Influenza , Gripe Aviar , Gripe Humana , Vacunas de Productos Inactivados , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Presentación de Antígeno/inmunología , Aves/inmunología , Aves/virología , Pruebas de Inhibición de Hemaglutinación , Humanos , Subtipo H5N1 del Virus de la Influenza A/inmunología , Subtipo H7N7 del Virus de la Influenza A/inmunología , Subtipo H9N2 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/inmunología , Gripe Aviar/inmunología , Gripe Aviar/prevención & control , Gripe Humana/inmunología , Gripe Humana/prevención & control , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología
20.
mBio ; 3(4): e00159-12, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22761392

RESUMEN

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.


Asunto(s)
Intestinos/virología , Organoides/virología , Infecciones por Rotavirus/virología , Rotavirus/fisiología , Células Madre/virología , Virología/métodos , Adulto , Diferenciación Celular , Células Cultivadas , Femenino , Humanos , Intestinos/citología , Masculino , Persona de Mediana Edad , Modelos Biológicos , Organoides/citología , Rotavirus/genética , Células Madre/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA