Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Virol ; 98(9): e0065624, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39136460

RESUMEN

The multifunctional tegument protein pUL21 of HSV-2 is phosphorylated in infected cells. We have identified two residues in the unstructured linker region of pUL21, serine 251 and serine 253, as phosphorylation sites. Both phosphorylation sites are absent in HSV-1 pUL21, which likely explains why phosphorylated pUL21 was not detected in cells infected with HSV-1. Cells infected with HSV-2 strain 186 viruses deficient in pUL21 phosphorylation exhibited reductions in both cell-cell spread of virus infection and virus replication. Defects in secondary envelopment of cytoplasmic nucleocapsids were also observed in cells infected with viruses deficient in pUL21 phosphorylation as well as in cells infected with multiple strains of HSV-2 and HSV-1 deleted for pUL21. These results confirm a role for HSV pUL21 in the secondary envelopment of cytoplasmic nucleocapsids and indicate that phosphorylation of HSV-2 pUL21 is required for this activity. Phosphorylation of pUL21 was substantially reduced in cells infected with HSV-2 strain 186 mutants lacking the viral serine/threonine kinase pUL13, indicating a requirement for pUL13 in pUL21 phosphorylation. IMPORTANCE: It is well known that post-translational modification of proteins by phosphorylation can regulate protein function. Here, we determined that phosphorylation of the multifunctional HSV-2 tegument protein pUL21 requires the viral serine/threonine kinase pUL13. In addition, we identified serine residues within HSV-2 pUL21 that can be phosphorylated. Phenotypic analysis of mutant HSV-2 strains with deficiencies in pUL21 phosphorylation revealed reductions in both cell-cell spread of virus infection and virus replication. Deficiencies in pUL21 phosphorylation also compromised the secondary envelopment of cytoplasmic nucleocapsids, a critical final step in the maturation of all herpes virions. Unlike HSV-2 pUL21, phosphorylation of HSV-1 pUL21 was not detected. This fundamental difference between HSV-2 and HSV-1 may underlie our previous observations that the requirements for pUL21 differ between HSV species.


Asunto(s)
Herpesvirus Humano 2 , Nucleocápside , Replicación Viral , Herpesvirus Humano 2/metabolismo , Herpesvirus Humano 2/genética , Herpesvirus Humano 2/fisiología , Fosforilación , Animales , Chlorocebus aethiops , Humanos , Células Vero , Nucleocápside/metabolismo , Herpesvirus Humano 1/fisiología , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 1/genética , Proteínas Virales/metabolismo , Proteínas Virales/genética , Citoplasma/metabolismo , Citoplasma/virología , Línea Celular , Proteínas Estructurales Virales/metabolismo , Proteínas Estructurales Virales/genética , Ensamble de Virus , Herpes Simple/virología , Herpes Simple/metabolismo
2.
PLoS Pathog ; 19(12): e1011832, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38039340

RESUMEN

After entry into cells, herpes simplex virus (HSV) nucleocapsids dock at nuclear pore complexes (NPCs) through which viral genomes are released into the nucleoplasm where viral gene expression, genome replication, and early steps in virion assembly take place. After their assembly, nucleocapsids are translocated to the cytoplasm for final virion maturation. Nascent cytoplasmic nucleocapsids are prevented from binding to NPCs and delivering their genomes to the nucleus from which they emerged, but how this is accomplished is not understood. Here we report that HSV pUL16 and pUL21 deletion mutants accumulate empty capsids at the cytoplasmic face of NPCs late in infection. Additionally, prior expression of pUL16 and pUL21 prevented incoming nucleocapsids from docking at NPCs, delivering their genomes to the nucleus and initiating viral gene expression. Both pUL16 and pUL21 localized to the nuclear envelope, placing them in an appropriate location to interfere with nucleocapsid/NPC interactions.


Asunto(s)
Herpes Simple , Herpesvirus Humano 1 , Humanos , Cápside/metabolismo , Poro Nuclear/metabolismo , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Nucleocápside/metabolismo
3.
PLoS Pathog ; 18(11): e1010969, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36374856

RESUMEN

During virion morphogenesis herpes simplex virus nucleocapsids transit from the nucleoplasm to the cytoplasm, through a process called nuclear egress, where the final stages of virion assembly occur. Coupled to nuclear egress is a poorly understood quality-control mechanism that preferentially selects genome-containing C-capsids, rather than A- and B-capsids that lack genomes, for transit to the cytoplasm. We and others have reported that cells infected with HSV strains deleted for the tegument protein pUL21 accumulate both empty A-capsids and C-capsids in the cytoplasm of infected cells. Quantitative microscopy experiments indicated that C-capsids were preferentially selected for envelopment at the inner nuclear membrane and that nuclear integrity remained intact in cells infected with pUL21 mutants, prompting alternative explanations for the accumulation of A-capsids in the cytoplasm. More A-capsids were also found in the nuclei of cells infected with pUL21 mutants compared to their wild type (WT) counterparts, suggesting pUL21 might be required for optimal genome packaging or genome retention within capsids. In support of this, more viral genomes were prematurely released into the cytoplasm during pUL21 mutant infection compared to WT infection and led to enhanced activation of cellular cytoplasmic DNA sensors. Mass spectrometry and western blot analysis of WT and pUL21 mutant capsids revealed an increased association of the known pUL21 binding protein, pUL16, with pUL21 mutant capsids, suggesting that premature and/or enhanced association of pUL16 with capsids might result in capsid destabilization. Further supporting this idea, deletion of pUL16 from a pUL21 mutant strain rescued genome retention within capsids. Taken together, these findings suggest that pUL21 regulates pUL16 addition to nuclear capsids and that premature, and/or, over-addition of pUL16 impairs HSV genome retention within capsids.


Asunto(s)
Herpes Simple , Herpesvirus Humano 1 , Humanos , Cápside/metabolismo , Herpesvirus Humano 1/genética , Ensamble de Virus/genética , Genoma Viral
4.
PLoS Pathog ; 17(8): e1009679, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34424922

RESUMEN

It is well established that the herpesvirus nuclear egress complex (NEC) has an intrinsic ability to deform membranes. During viral infection, the membrane-deformation activity of the NEC must be precisely regulated to ensure efficient nuclear egress of capsids. One viral protein known to regulate herpes simplex virus type 2 (HSV-2) NEC activity is the tegument protein pUL21. Cells infected with an HSV-2 mutant lacking pUL21 (ΔUL21) produced a slower migrating species of the viral serine/threonine kinase pUs3 that was shown to be a hyperphosphorylated form of the enzyme. Investigation of the pUs3 substrate profile in ΔUL21-infected cells revealed a prominent band with a molecular weight consistent with that of the NEC components pUL31 and pUL34. Phosphatase sensitivity and retarded mobility in phos-tag SDS-PAGE confirmed that both pUL31 and pUL34 were hyperphosphorylated by pUs3 in the absence of pUL21. To gain insight into the consequences of increased phosphorylation of NEC components, the architecture of the nuclear envelope in cells producing the HSV-2 NEC in the presence or absence of pUs3 was examined. In cells with robust NEC production, invaginations of the inner nuclear membrane were observed that contained budded vesicles of uniform size. By contrast, nuclear envelope deformations protruding outwards from the nucleus, were observed when pUs3 was included in transfections with the HSV-2 NEC. Finally, when pUL21 was included in transfections with the HSV-2 NEC and pUs3, decreased phosphorylation of NEC components was observed in comparison to transfections lacking pUL21. These results demonstrate that pUL21 influences the phosphorylation status of pUs3 and the HSV-2 NEC and that this has consequences for the architecture of the nuclear envelope.


Asunto(s)
Herpes Simple/patología , Herpesvirus Humano 2/fisiología , Membrana Nuclear/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Virales/metabolismo , Liberación del Virus , Animales , Cápside/fisiología , Núcleo Celular/genética , Núcleo Celular/metabolismo , Chlorocebus aethiops , Células HeLa , Herpes Simple/metabolismo , Herpes Simple/virología , Humanos , Membrana Nuclear/metabolismo , Membrana Nuclear/virología , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Células Vero , Proteínas Virales/genética , Ensamble de Virus
5.
J Virol ; 94(13)2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32321804

RESUMEN

Viral proteins pUL16 and pUL21 are required for efficient nuclear egress of herpes simplex virus 2 capsids. To better understand the role of these proteins in nuclear egress, we established whether nuclear egress complex (NEC) distribution and/or function was altered in the absence of either pUL16 or pUL21. NEC distribution in cells infected with pUL16-deficient viruses was indistinguishable from that observed in cells infected with wild-type viruses. In contrast, NEC distribution was aberrant in cells infected with pUL21-deficient virus and, instead, showed some similarity to the aberrant NEC distribution pattern observed in cells infected with pUs3-deficient virus. These results indicated that pUL16 plays a role in nuclear egress that is distinct from that of pUL21 and pUs3. Higher-resolution examination of nuclear envelope ultrastructure in cells infected with pUL21-deficient viruses by transmission electron microscopy showed different types of nuclear envelope perturbations, including some that were not observed in cells infected with pUs3 deficient virus. The formation of the nuclear envelope perturbations observed in pUL21-deficient virus infections was dependent on a functional NEC, revealing a novel role for pUL21 in regulating NEC activity. The results of comparisons of nuclear envelope ultrastructure in cells infected with viruses lacking pUs3, pUL16, or both pUs3 and pUL16 were consistent with a role for pUL16 in advance of primary capsid envelopment and shed new light on how pUs3 functions in nuclear egress.IMPORTANCE The membrane deformation activity of the herpesvirus nuclear egress complex (NEC) allows capsids to transit through both nuclear membranes into the cytoplasm. NEC activity must be precisely controlled during viral infection, and yet our knowledge of how NEC activity is controlled is incomplete. To determine how pUL16 and pUL21, two viral proteins required for nuclear egress of herpes simplex virus 2, function in nuclear egress, we examined how the lack of each protein impacted NEC distribution. These analyses revealed a function of pUL16 in nuclear egress distinct from that of pUL21, uncovered a novel role for pUL21 in regulating NEC activity, and shed new light on how a viral kinase, pUs3, regulates nuclear egress. Nuclear egress of capsids is required for all herpesviruses. A complete understanding of all aspects of nuclear egress, including how viral NEC activity is controlled, may yield strategies to disrupt this process and aid the development of herpes-specific antiviral therapies.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Virales/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Animales , Cápside/metabolismo , Proteínas de la Cápside/metabolismo , Núcleo Celular/virología , Chlorocebus aethiops , Fibroblastos , Células HeLa , Herpes Simple/virología , Infecciones por Herpesviridae/metabolismo , Herpesvirus Humano 1/fisiología , Herpesvirus Humano 2/fisiología , Humanos , Ratones , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Simplexvirus/metabolismo , Simplexvirus/patogenicidad , Células Vero , Proteínas Virales/fisiología , Proteínas Reguladoras y Accesorias Virales/fisiología , Virión/metabolismo , Ensamble de Virus , Liberación del Virus/fisiología , Replicación Viral
6.
J Biol Chem ; 293(45): 17631-17645, 2018 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-30242126

RESUMEN

Upon repeated exposure to endotoxin or lipopolysaccharide (LPS), myeloid cells enter a refractory state called endotoxin tolerance as a homeostatic mechanism. In innate immune cells, LPS is recognized by co-receptors Toll-like receptor 4 (TLR4) and CD-14 to initiate an inflammatory response for subsequent cytokine production. One such cytokine, interleukin (IL)-27, is produced by myeloid cells in response to bacterial infection. In monocytes, IL-27 has proinflammatory functions such as up-regulating TLR4 expression for enhanced LPS-mediated cytokine production; alternatively, IL-27 induces inhibitory functions in activated macrophages. This study investigated the effects of IL-27 on the induction of endotoxin tolerance in models of human monocytes compared with macrophages. Our data demonstrate that IL-27 inhibits endotoxin tolerance by up-regulating cell surface TLR4 expression and soluble CD14 production to mediate stability of the surface LPS-TLR4-CD14 complex in THP-1 cells. In contrast, elevated basal expression of membrane-bound CD14 in phorbol 12-myristate 13-acetate (PMA)-THP-1 cells, primary monocytes, and primary macrophages may promote CD14-mediated endocytosis and be responsible for the preservation of an endotoxin-tolerized state in the presence of IL-27. Overall, the efficacy of IL-27 in inhibiting endotoxin tolerance in human THP-1 monocytes and PMA-THP-1 macrophages is affected by membrane-bound and soluble CD14 expression.


Asunto(s)
Tolerancia Inmunológica/efectos de los fármacos , Interleucinas/inmunología , Receptores de Lipopolisacáridos/inmunología , Lipopolisacáridos/toxicidad , Macrófagos/inmunología , Modelos Inmunológicos , Monocitos/inmunología , Endocitosis/efectos de los fármacos , Endocitosis/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Humanos , Activación de Macrófagos/efectos de los fármacos , Células THP-1 , Acetato de Tetradecanoilforbol/farmacología , Receptor Toll-Like 4/inmunología
7.
Viruses ; 10(5)2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29762484

RESUMEN

Studies from multiple laboratories using different strains or species of herpes simplex virus (HSV) with deletions in UL21 have yielded conflicting results regarding the necessity of pUL21 in HSV infection. To resolve this discrepancy, we utilized CRISPR/Cas9 mutagenesis to isolate pUL21 deficient viruses in multiple HSV backgrounds, and performed a side-by-side comparison of the cell-to-cell spread and replication phenotypes of these viruses. These analyses confirmed previous studies implicating the involvement of pUL21 in cell-to-cell spread of HSV. Cell-to-cell spread of HSV-2 was more greatly affected by the lack of pUL21 than HSV-1, and strain-specific differences in the requirement for pUL21 in cell-to-cell spread were also noted. HSV-2 strain 186 lacking pUL21 was particularly crippled in both cell-to-cell spread and viral replication in non-complementing cells, in comparison to other HSV strains lacking pUL21, suggesting that the strict requirement for pUL21 by strain 186 may not be representative of the HSV-2 species as a whole. This work highlights CRISPR/Cas9 technology as a useful tool for rapidly constructing deletion mutants of alphaherpesviruses, regardless of background strain, and should find great utility whenever strain-specific differences need to be investigated.


Asunto(s)
Sistemas CRISPR-Cas , Herpes Simple/virología , Herpesvirus Humano 1/fisiología , Herpesvirus Humano 2/fisiología , Mutagénesis , Proteínas Virales/metabolismo , Replicación Viral , Animales , Línea Celular , Genes Virales/genética , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/crecimiento & desarrollo , Herpesvirus Humano 2/genética , Herpesvirus Humano 2/crecimiento & desarrollo , Humanos , Eliminación de Secuencia , Ensayo de Placa Viral , Proteínas Virales/genética
8.
J Virol ; 92(13)2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29669832

RESUMEN

Orthologs of the herpes simplex virus (HSV) UL16 gene are conserved throughout the Herpesviridae Because of this conservation, one might expect that the proteins perform similar functions for all herpesviruses. Previous studies on a UL16-null mutant derived from HSV-2 strain 186 revealed a roughly 100-fold replication defect and a critical role for UL16 in the nuclear egress of capsids. These findings were in stark contrast to what has been observed with UL16 mutants of HSV-1 and pseudorabies virus, where roughly 10-fold replication deficiencies that were accompanied by defects in the secondary envelopment of cytoplasmic capsids were reported. One possible explanation for this discrepancy is that HSV-2 strain 186 is not representative of the HSV-2 species. To address this possibility, multiple UL16-null mutants were constructed in multiple HSV-2 and HSV-1 strains by CRISPR/Cas9 mutagenesis, and their phenotypes were characterized side by side. This analysis showed that all the HSV-2 UL16 mutants had 50- to 100-fold replication deficiencies that were accompanied by defects in the nuclear egress of capsids, as well as defects in the secondary envelopment of cytoplasmic capsids. By contrast, most HSV-1 UL16 mutants had 10-fold replication deficiencies that were accompanied by defects in secondary envelopment of cytoplasmic capsids. These findings indicated that UL16 has HSV species-specific functions. Interestingly, HSV-1 UL16 could promote the nuclear egress of HSV-2 UL16-null strains, suggesting that, unlike HSV-1, HSV-2 lacks an activity that can promote nuclear egress in the absence of UL16.IMPORTANCE HSV-2 and HSV-1 are important human pathogens that cause distinct diseases in their hosts. A complete understanding of the morphogenesis of these viruses is expected to reveal vulnerabilities that can be exploited in the treatment of HSV disease. UL16 is a virion structural component that is conserved throughout the Herpesviridae and functions in virus morphogenesis; however, previous studies have suggested different roles for UL16 in the morphogenesis of HSV-2 and HSV-1. This study sought to resolve this apparent discrepancy by analyzing multiple UL16 mutant viruses derived from multiple strains of HSV-2 and HSV-1. The data indicate that UL16 has HSV species-specific functions, as HSV-2 has a requirement for UL16 in the escape of capsids from the nucleus whereas both HSV-2 and HSV-1 require UL16 for final envelopment of capsids at cytoplasmic membranes.


Asunto(s)
Herpes Simple/virología , Herpesvirus Humano 1/fisiología , Herpesvirus Humano 2/fisiología , Mutación , Proteínas Virales/metabolismo , Replicación Viral , Animales , Chlorocebus aethiops , Humanos , Especificidad de la Especie , Células Vero , Proteínas Virales/genética
9.
Sci Rep ; 7(1): 1882, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28507315

RESUMEN

The herpes simplex virus (HSV) UL31 gene encodes a conserved member of the herpesvirus nuclear egress complex that not only functions in the egress of DNA containing capsids from the nucleus, but is also required for optimal replication of viral DNA and its packaging into capsids. Here we report that the UL31 protein from HSV-2 can be recruited to sites of DNA damage by sequences found in its N-terminus. The N-terminus of UL31 contains sequences resembling a poly (ADP-ribose) (PAR) binding motif suggesting that PAR interactions might mediate UL31 recruitment to damaged DNA. Whereas PAR polymerase inhibition prevented UL31 recruitment to damaged DNA, inhibition of signaling through the ataxia telangiectasia mutated DNA damage response pathway had no effect. These findings were further supported by experiments demonstrating direct and specific interaction between HSV-2 UL31 and PAR using purified components. This study reveals a previously unrecognized function for UL31 and may suggest that the recognition of PAR by UL31 is coupled to the nuclear egress of herpesvirus capsids, influences viral DNA replication and packaging, or possibly modulates the DNA damage response mounted by virally infected cells.


Asunto(s)
Daño del ADN , Herpesvirus Humano 2/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Proteínas Virales/metabolismo , Evolución Biológica , Línea Celular , Daño del ADN/efectos de la radiación , Expresión Génica , Genes Reporteros , Herpesvirus Humano 2/genética , Humanos , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Unión Proteica , Transporte de Proteínas , Proteínas Virales/genética
10.
J Leukoc Biol ; 102(1): 83-94, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28377398

RESUMEN

IL-27 bridges innate and adaptive immunity by modulating cytokine production from myeloid cells and regulating Th cell differentiation. During bacterial infection, TLR4 triggering by LPS induces IL-27 production by monocytes and macrophages. We have previously shown that IL-27 can prime monocytes for LPS responsiveness by enhancing TLR4 expression and intracellular signaling. If unregulated, this could result in damaging inflammation, whereas on the other hand, this may also provide greater responses by inflammatory processes induced in response to bacterial pathogens. A key process in fine-tuning inflammatory responses is activation of the inflammasome, which ultimately results in IL-1ß production. Herein, we investigated the molecular mechanisms by which IL-27 modulates LPS-induced IL-1ß secretion in monocytes and macrophages. We found that when delivered simultaneously with LPS, IL-27 augments activation of caspase-1 and subsequent release of IL-1ß. Furthermore, we determined that IL-27 primes cells for enhanced IL-1ß production by up-regulating surface expression of TLR4 and P2X purinoceptor 7 (P2X7) for enhanced LPS and ATP signaling, respectively. These findings provide new evidence that IL-27 plays an important role in the proinflammatory capacity of monocytes and macrophages via enhancing IL-1ß secretion levels triggered by dual LPS-ATP stimulation.


Asunto(s)
Interleucina-1beta/inmunología , Interleucinas/inmunología , Lipopolisacáridos/farmacología , Macrófagos/inmunología , Monocitos/inmunología , Transducción de Señal/efectos de los fármacos , Adenosina Trifosfato/farmacología , Animales , Caspasa 1/genética , Caspasa 1/inmunología , Línea Celular Tumoral , Humanos , Interleucina-1beta/genética , Interleucinas/genética , Ratones , Ratones Noqueados , Receptores Purinérgicos P2X7/genética , Receptores Purinérgicos P2X7/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...