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













Base de datos
Intervalo de año de publicación
1.
Virus Res ; 345: 199379, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38643859

RESUMEN

Although all herpesviruses utilize a highly conserved replication machinery to amplify their viral genomes, different members may have unique strategies to modulate the assembly of their replication components. Herein, we characterize the subcellular localization of seven essential replication proteins of varicella-zoster virus (VZV) and show that several viral replication enzymes such as the DNA polymerase subunit ORF28, when expressed alone, are localized in the cytoplasm. The nuclear import of ORF28 can be mediated by the viral DNA polymerase processivity factor ORF16. Besides, ORF16 could markedly enhance the protein abundance of ORF28. Noteworthily, an ORF16 mutant that is defective in nuclear transport still retained the ability to enhance ORF28 abundance. The low abundance of ORF28 in transfected cells was due to its rapid degradation mediated by the ubiquitin-proteasome system. We additionally reveal that radicicol, an inhibitor of the chaperone Hsp90, could disrupt the interaction between ORF16 and ORF28, thereby affecting the nuclear entry and protein abundance of ORF28. Collectively, our findings imply that the cytoplasmic retention and rapid degradation of ORF28 may be a key regulatory mechanism for VZV to prevent untimely viral DNA replication, and suggest that Hsp90 is required for the interaction between ORF16 and ORF28.


Asunto(s)
Transporte Activo de Núcleo Celular , ADN Polimerasa Dirigida por ADN , Herpesvirus Humano 3 , Proteínas Virales , Replicación Viral , Herpesvirus Humano 3/genética , Herpesvirus Humano 3/metabolismo , Humanos , Proteínas Virales/metabolismo , Proteínas Virales/genética , ADN Polimerasa Dirigida por ADN/metabolismo , ADN Polimerasa Dirigida por ADN/genética , Núcleo Celular/metabolismo , Núcleo Celular/virología , Citoplasma/metabolismo , Citoplasma/virología , Línea Celular , Replicación del ADN
2.
Microbiol Spectr ; 11(6): e0225423, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-37874136

RESUMEN

IMPORTANCE: Eukaryotic DNA replication is a highly regulated process that requires multiple replication enzymes assembled onto DNA replication origins. Due to the complexity of the cell's DNA replication machinery, most of what we know about cellular DNA replication has come from the study of viral systems. Herein, we focus our study on the assembly of the Kaposi's sarcoma-associated herpesvirus core replication complex and propose a pairwise protein-protein interaction network of six highly conserved viral core replication proteins. A detailed understanding of the interaction and assembly of the viral core replication proteins may provide opportunities to develop new strategies against viral propagation.


Asunto(s)
Herpesvirus Humano 8 , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Proteínas Virales/genética , Replicación del ADN
3.
Int J Mol Sci ; 23(15)2022 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-35955808

RESUMEN

The Kaposi's sarcoma-associated herpesvirus (KSHV)-encoded ORF50 protein is a potent transcriptional activator essential for triggering KSHV lytic reactivation. Despite extensive studies, little is known about whether ORF50 possesses the ability to repress gene expression or has an antagonistic action to cellular transcription factors. Previously, we demonstrated that human oncoprotein MDM2 can promote the degradation of ORF50 protein. Herein, we show that abundant ORF50 expression in cells can conversely downregulate MDM2 expression via repressing both the upstream (P1) and internal (P2) promoters of the MDM2 gene. Deletion analysis of the MDM2 P1 promoter revealed that there were two ORF50-dependent negative response elements located from -102 to -63 and from -39 to +1, which contain Sp1-binding sites. For the MDM2 P2 promoter, the ORF50-dependent negative response element was identified in the region from -110 to -25, which is coincident with the location of two known p53-binding sites. Importantly, we further demonstrated that overexpression of Sp1 or p53 in cells indeed upregulated MDM2 expression; however, coexpression with ORF50 protein remarkably reduced the Sp1- or p53-mediated MDM2 upregulation. Collectively, our findings propose a reciprocal negative regulation between ORF50 and MDM2 and uncover that ORF50 decreases MDM2 expression through repressing Sp1- and p53-mediated transactivation.


Asunto(s)
Herpesvirus Humano 8 , Regulación Viral de la Expresión Génica , Herpesvirus Humano 8/genética , Humanos , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Elementos de Respuesta , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Virales
4.
Microbiol Spectr ; 10(1): e0146421, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35019687

RESUMEN

The open reading frame 50 (ORF50) protein of Kaposi's sarcoma-associated herpesvirus (KSHV) is the master regulator essential for initiating the viral lytic cycle. Previously, we have demonstrated that the ORF50 protein can cooperate with Sp3 to synergistically activate a set of viral and cellular gene promoters through highly conserved ORF50-responsive elements that harbor a Sp3-binding motif. Herein, we show that Sp3 undergoes proteolytic cleavage during the viral lytic cycle, and the cleavage of Sp3 is dependent on caspase activation. Since similar cleavage patterns of Sp3 could be detected in both KSHV-positive and KSHV-negative lymphoma cells undergoing apoptosis, the proteolytic cleavage of Sp3 could be a common event during apoptosis. Mutational analysis identifies 12 caspase cleavage sites in Sp3, which are situated at the aspartate (D) positions D17, D19, D180, D273, D275, D293, D304 (or D307), D326, D344, D530, D543, and D565. Importantly, we noticed that three stable Sp3 C-terminal fragments generated through cleavage at D530, D543, or D565 encompass an intact DNA-binding domain. Like the full-length Sp3, the C-terminal fragments of Sp3 could still retain the ability to cooperate with ORF50 protein to activate specific viral and cellular gene promoters synergistically. Collectively, our findings suggest that despite the proteolytic cleavage of Sp3 under apoptotic conditions, the resultant Sp3 fragments may retain biological activities important for the viral lytic cycle or for cellular apoptosis. IMPORTANCE The ORF50 protein of Kaposi's sarcoma-associated herpesvirus (KSHV) is the key viral protein that controls the switch from latency to lytic reactivation. It is a potent transactivator that can activate target gene promoters via interacting with other cellular DNA-binding transcription factors, such as Sp3. In this report, we show that Sp3 is proteolytically cleaved during the viral lytic cycle, and up to 12 caspase cleavage sites are identified in Sp3. Despite the proteolytic cleavage of Sp3, several resulting C-terminal fragments that have intact zinc-finger DNA-binding domains still retain substantial influence in the synergy with ORF50 to activate specific gene promoters. Overall, our studies elucidate the caspase-mediated cleavage of Sp3 and uncover how ORF50 utilizes the cleavage fragments of Sp3 to transactivate specific viral and cellular gene promoters.


Asunto(s)
Caspasas/metabolismo , Infecciones por Herpesviridae/metabolismo , Herpesvirus Humano 8/fisiología , Factor de Transcripción Sp3/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Apoptosis , Caspasas/genética , Regulación Viral de la Expresión Génica , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/fisiopatología , Infecciones por Herpesviridae/virología , Herpesvirus Humano 8/genética , Interacciones Huésped-Patógeno , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Linfoma/genética , Linfoma/metabolismo , Linfoma/fisiopatología , Linfoma/virología , Alineación de Secuencia , Factor de Transcripción Sp3/química , Factor de Transcripción Sp3/genética , Transactivadores/genética , Transactivadores/metabolismo , Latencia del Virus
5.
Microbiol Spectr ; 9(1): e0042221, 2021 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-34431704

RESUMEN

The human norovirus (HuNV)-encoded nucleoside-triphosphatase (NTPase) is a multifunctional protein critically involved in viral replication and pathogenesis. Previously, we have shown that the viral NTPase is capable of forming vesicle clusters in cells, interacting with other viral proteins such as P22, and promoting cellular apoptosis. Herein, we demonstrate that NTPase-associated vesicle clusters correspond to lipid droplets (LDs) wrapped by the viral protein and show that NTPase-induced apoptosis is mediated through both caspase-8- and caspase-9-dependent pathways. Deletion analysis revealed that the N-terminal 179-amino-acid (aa) region of NTPase encompasses two LD-targeting motifs (designated LTM-1 and LTM-2), two apoptosis-inducing motifs, and multiple regulatory regions. Interestingly, the identified LTM-1 and LTM-2, which are located from aa 1 to 50 and from aa 51 to 90, respectively, overlap with the two apoptosis-inducing motifs. Although there was no positive correlation between the extent of LD localization and the degree of cellular apoptosis for NTPase mutants, we noticed that mutant proteins defective in LD-targeting ability could not induce cellular apoptosis. In addition to LD targeting, the amphipathic LTM-1 and LTM-2 motifs could have the potential to direct fusion proteins to the endoplasmic reticulum (ER). Furthermore, we found that the LTM-1 motif is a P22-interacting motif. However, P22 functionally augmented the proapoptotic activity of the LTM-2 fusion protein but not the LTM-1 fusion protein. Overall, our findings propose that NTPase may participate in multiple cellular processes through binding to LDs or to the ER via its N-terminal amphipathic helix motifs. IMPORTANCE Human noroviruses (HuNVs) are the major agent of global gastroenteritis outbreaks. However, due to the lack of an efficient cell culture system for HuNV propagation, functions of the viral-encoded proteins in host cells are still poorly understood. In the current study, we present that the viral NTPase is a lipid droplet (LD)-associated protein, and we identify two LD-targeting motifs, LTM-1 and LTM-2, in its N-terminal domain. In particular, the identified LTM-1 and LTM-2 motifs, which contain a hydrophobic region and an amphipathic helix, are also capable of delivering the fusion protein to the endoplasmic reticulum (ER), promoting cellular apoptosis, and physically or functionally associating with another viral protein P22. Since LDs and the ER have been linked to several biological functions in cells, our study therefore proposes that the norovirus NTPase may utilize LDs or the ER as replication platforms to benefit viral replication and pathogenesis.


Asunto(s)
Gotas Lipídicas/metabolismo , Norovirus/enzimología , Nucleósido-Trifosfatasa/aislamiento & purificación , Proteínas Virales/metabolismo , Apoptosis , Retículo Endoplásmico/metabolismo , Gastroenteritis , Humanos , Norovirus/genética , Nucleósido-Trifosfatasa/genética , Replicación Viral
6.
Int J Mol Sci ; 22(8)2021 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-33919712

RESUMEN

The unfolded protein response (UPR) is an intracellular signaling pathway essential for alleviating the endoplasmic reticulum (ER) stress. To support the productive infection, many viruses are known to use different strategies to manipulate the UPR signaling network. However, it remains largely unclear whether the UPR signaling pathways are modulated in the lytic cycle of Epstein-Barr virus (EBV), a widely distributed human pathogen. Herein, we show that the expression of GRP78, a central UPR regulator, is up-regulated during the EBV lytic cycle. Our data further revealed that knockdown of GRP78 in EBV-infected cell lines did not substantially affect lytic gene expression; however, GRP78 knockdown in these cells markedly reduced the production of virus particles. Importantly, we identified that the early lytic protein BMLF1 is the key regulator critically contributing to the activation of the grp78 gene promoter. Mechanistically, we found that BMLF1 can trigger the proteolytic cleavage and activation of the UPR senor ATF6, which then transcriptionally activates the grp78 promoter through the ER stress response elements. Our findings therefore provide evidence for the connection between the EBV lytic cycle and the UPR, and implicate that the BMLF1-mediated ATF6 activation may play critical roles in EBV lytic replication.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Proteínas de Choque Térmico/genética , Fosfoproteínas/metabolismo , Transactivadores/metabolismo , Regulación hacia Arriba , Secuencia de Bases , Línea Celular Tumoral , Núcleo Celular/metabolismo , ADN Viral/biosíntesis , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico , Endorribonucleasas/metabolismo , Regulación Viral de la Expresión Génica , Células HEK293 , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/fisiología , Humanos , Modelos Biológicos , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas , Transducción de Señal , Activación Transcripcional/genética , Respuesta de Proteína Desplegada , Regulación hacia Arriba/genética , eIF-2 Quinasa/metabolismo
7.
J Virol ; 94(18)2020 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-32641483

RESUMEN

The Kaposi's sarcoma-associated herpesvirus (KSHV)-encoded open reading frame 50 (ORF50) protein is the key transactivator responsible for the latent-to-lytic switch. Here, we investigated the transcriptional activation of the ORF56 gene (encoding a primase protein) by ORF50 and successfully identified an ORF50-responsive element located in the promoter region between positions -97 and -44 (designated 56p-RE). This 56p-RE element contains a noncanonical RBP-Jκ-binding sequence and a nonconsensus Sp1/Sp3-binding sequence. Electrophoretic mobility shift assays revealed that RBP-Jκ, Sp3, and ORF50 could form stable complexes on the 56p-RE element. Importantly, transient-reporter analysis showed that Sp3, but not RBP-Jκ or Sp1, acts in synergy with ORF50 to activate the 56p-RE-containing reporter construct, and the synergy mainly depends on the Sp1/Sp3-binding region of the 56p-RE element. Sequence similarity searches revealed that the promoters for ORF21 (thymidine kinase), ORF60 (ribonucleotide reductase, small subunit), and cellular interleukin-10 (IL-10) contain a sequence motif similar to the Sp1/Sp3-binding region of the 56p-RE element, and we found that these promoters could also be synergistically activated by ORF50 and Sp3 via the conserved motifs. Noteworthily, the conversion of the Sp1/Sp3-binding sequence of the 56p-RE element into a consensus high-affinity Sp-binding sequence completely lost the synergistic response to ORF50 and Sp3. Moreover, transcriptional synergy could not be detected through other ORF50-responsive elements from the viral PAN, K12, ORF57, and K6 promoters. Collectively, the results of our study demonstrate that ORF50 and Sp3 can act in synergy on the transcription of specific gene promoters, and we find a novel conserved cis-acting motif in these promoters essential for transcriptional synergy.IMPORTANCE Despite the critical role of ORF50 in the KSHV latent-to-lytic switch, the molecular mechanism by which ORF50 activates its downstream target genes, especially those that encode the viral DNA replication enzymes, is not yet fully understood. Here, we find that ORF50 can cooperate with Sp3 to synergistically activate promoters of the viral ORF56 (primase), ORF21 (thymidine kinase), and ORF60 (ribonucleotide reductase) genes via similar Sp1/Sp3-binding motifs. Additionally, the same synergistic effect can be seen on the promoter of the cellular IL-10 gene. Overall, our data reveal an important role for Sp3 in ORF50-mediated transactivation, and we propose a new subclass of ORF50-responsive elements in viral and cellular promoters.


Asunto(s)
Herpesvirus Humano 8/genética , Proteínas Inmediatas-Precoces/genética , Regiones Promotoras Genéticas , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp3/genética , Transactivadores/genética , Transcripción Genética , Animales , Sitios de Unión , Línea Celular , Línea Celular Tumoral , Células Clonales , Fibroblastos/virología , Regulación de la Expresión Génica , Células HEK293 , Herpesvirus Humano 8/metabolismo , Interacciones Huésped-Patógeno/genética , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Linfocitos/virología , Ratones , Unión Proteica , Elementos de Respuesta , Transducción de Señal , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción Sp3/metabolismo , Transactivadores/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Reguladoras y Accesorias Virales/metabolismo
8.
Virus Res ; 256: 76-89, 2018 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-30096410

RESUMEN

The BKRF2, BKRF3 and BKRF4 genes of Epstein-Barr virus (EBV) are located close together in the viral genome, which encode glycoprotein L, uracil-DNA glycosylase and a tegument protein, respectively. Here, we demonstrate that the BKRF2 gene behaves as a true-late lytic gene, whereas the BKRF3 and BKRF4 genes belong to the early lytic gene family. Our results further reveal that both BKRF3 and BKRF4 promoters are new synergistic targets of Zta and Rta, two EBV latent-to-lytic switch transactivators. Multiple Rta- and Zta-responsive elements within the BKRF3 and BKRF4 promoters were identified and characterized experimentally. Importantly, we show that DNA methylation is absolutely required for activation of the BKRF4 promoter by Zta alone or in combination with Rta. Moreover, we find that sodium butyrate, an inducing agent of EBV reactivation, is capable of activating the BKRF4 promoter through a mechanism independent of Zta and Rta. Overall, our studies highlight the complexity of transcriptional regulation of lytic genes within the BKRF2-BKRF3-BKRF4 gene locus.


Asunto(s)
Regulación Viral de la Expresión Génica , Herpesvirus Humano 4/crecimiento & desarrollo , Herpesvirus Humano 4/genética , Glicoproteínas de Membrana/genética , Chaperonas Moleculares/genética , Uracil-ADN Glicosidasa/genética , Proteínas Virales/genética , Metilación de ADN , ADN Viral/metabolismo , Perfilación de la Expresión Génica , Proteínas Inmediatas-Precoces/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Transactivadores/metabolismo
9.
J Virol ; 92(5)2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29212938

RESUMEN

The genotype II.4 (GII.4) variants of human noroviruses (HuNVs) are recognized as the major agent of global gastroenteritis outbreaks. Due to the lack of an efficient cell culture system for HuNV propagation, the exact roles of HuNV-encoded nonstructural proteins (including Nterm, NTPase, P22, VPg, Pro, and RdRp) in viral replication or pathogenesis have not yet been fully understood. Here, we report the molecular characterization of the GII.4 HuNV-encoded NTPase (designated GII-NTPase). Results from our studies showed that GII-NTPase forms vesicular or nonvesicular textures in the cell cytoplasm, and the nonvesicular fraction of GII-NTPase significantly localizes to the endoplasmic reticulum (ER) or mitochondria. Deletion analysis revealed that the N-terminal 179-amino-acid (aa) region of GII-NTPase is required for vesicle formation and for ER colocalization, whereas the C-terminal region is involved in mitochondrial colocalization. In particular, two mitochondrion-targeting domains were identified in the C-terminal region of GII-NTPase which perfectly colocalized with mitochondria when the N-terminal region of GII-NTPase was deleted. However, the corresponding C-terminal portions of NTPase derived from the GI HuNV did not show mitochondrial colocalization. We also found that GII-NTPase physically interacts with itself as well as with Nterm and P22, but not VPg, Pro, and RdRp, in cells. The Nterm- and P22-interacting region was mapped to the N-terminal 179-aa region of GII-NTPase, whereas the self-assembly of GII-NTPase could be achieved via a head-to-head, tail-to-tail, or head-to-tail configuration. More importantly, we demonstrate that GII-NTPase possesses a proapoptotic activity, which can be further enhanced by coexpression with Nterm or P22.IMPORTANCE Despite the importance of human norovirus GII.4 variants in global gastroenteritis outbreaks, the basic biological functions of the viral nonstructural proteins in cells remain rarely investigated. In this report, we focus our studies on characteristics of the GII.4 norovirus-encoded NTPase (GII-NTPase). We unexpectedly find that GII-NTPase can perfectly colocalize with mitochondria after its N-terminal region is deleted. However, such a phenomenon is not observed for NTPase encoded by a GI strain. We further reveal that the N-terminal 179-aa region of GII-NTPase is sufficient to mediate (i) vesicle formation, (ii) ER colocalization, (iii) the interaction with two other nonstructural proteins, including Nterm and P22, (iv) the formation of homodimers or homo-oligomers, and (v) the induction of cell apoptosis. Taken together, our findings emphasize that the virus-encoded NTPase must have multiple activities during viral replication or pathogenesis; however, these activities may vary somewhat among different genogroups.


Asunto(s)
Norovirus/enzimología , Norovirus/genética , Nucleósido-Trifosfatasa/genética , Nucleósido-Trifosfatasa/metabolismo , Secuencia de Aminoácidos , Apoptosis , Infecciones por Caliciviridae/virología , Mapeo Cromosómico , Citoplasma/metabolismo , Brotes de Enfermedades , Retículo Endoplásmico/metabolismo , Gastroenteritis/virología , Genotipo , Células HEK293 , Humanos , Mitocondrias/metabolismo , Norovirus/clasificación , Norovirus/patogenicidad , Nucleósido-Trifosfatasa/química , Nucleósido-Trifosfatasa/inmunología , Dominios y Motivos de Interacción de Proteínas , Alineación de Secuencia , Eliminación de Secuencia , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Replicación Viral
10.
Oncotarget ; 8(46): 80595-80611, 2017 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-29113328

RESUMEN

Patients with diabetes are generally prone to pathogen infection and tumor progression. Here, we investigated the potential association between diabetes and Kaposi's sarcoma (KS), a tumor linked to infection with Kaposi's sarcoma-associated herpesvirus (KSHV). By using Taiwan's National Health Insurance Research Database, we found that diabetes is statistically associated with increased risk of KS in a case-control study. Since a high level of blood sugar is the hallmark of diabetes, we determined whether high glucose promotes both KSHV reactivation and infection, which are crucial for KS pathogenesis. Our results showed that high glucose significantly increases lytic reactivation of KSHV but not Epstein-Barr virus, another related human oncogenic gammaherpesvirus, in latently infected cells. Activation of the transcription factor AP1 by high glucose is critically required for the onset of KSHV lytic reactivation. We also demonstrated that high glucose enhances susceptibility of various target cells to KSHV infection. Particularly, in endothelial and epithelial cells, levels of specific cellular receptors for KSHV entry, including integrin α3ß1 and xCT/CD98, are elevated under high glucose conditions, which correlate with the enhanced cell susceptibility to infection. Taken together, our studies implicate that the high-glucose microenvironment may be an important predisposing factor for KS development.

11.
J Virol ; 91(19)2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28701396

RESUMEN

The switch of Kaposi's sarcoma-associated herpesvirus (KSHV) from latency to lytic replication is a key event for viral dissemination and pathogenesis. MLN4924, a novel neddylation inhibitor, reportedly causes the onset of KSHV reactivation but impairs later phases of the viral lytic program in infected cells. Thus far, the molecular mechanism involved in the modulation of the KSHV lytic cycle by MLN4924 is not yet fully understood. Here, we confirmed that treatment of different KSHV-infected primary effusion lymphoma (PEL) cell lines with MLN4924 substantially induces viral lytic protein expression. Due to the key role of the virally encoded ORF50 protein in the latent-to-lytic switch, we investigated its transcriptional regulation by MLN4924. We found that MLN4924 activates the ORF50 promoter (ORF50p) in KSHV-positive cells (but not in KSHV-negative cells), and the RBP-Jκ-binding elements within the promoter are critically required for MLN4924 responsiveness. In KSHV-negative cells, reactivation of the ORF50 promoter by MLN4924 requires the presence of the latency-associated nuclear antigen (LANA). Under such a condition, LANA acts as a repressor to block the ORF50p activity, whereas MLN4924 treatment relieves LANA-mediated repression. Importantly, we showed that LANA is a neddylated protein and can be deneddylated by MLN4924. On the other hand, we revealed that MLN4924 exhibits concentration-dependent biphasic effects on 12-O-tetradecanoylphorbol-13-acetate (TPA)- or sodium butyrate (SB)-induced viral reactivation in PEL cell lines. In other words, low concentrations of MLN4924 promote activation of TPA- or SB-mediated viral reactivation, whereas high concentrations of MLN4924, conversely, inhibit the progression of TPA- or SB-mediated viral lytic program.IMPORTANCE MLN4924 is a neddylation (NEDD8 modification) inhibitor, which currently acts as an anti-cancer drug in clinical trials. Although MLN4924 has been reported to trigger KSHV reactivation, many aspects regarding the action of MLN4924 in regulating the KSHV lytic cycle are not fully understood. Since the KSHV ORF50 protein is the key regulator of viral lytic reactivation, we focus on its transcriptional regulation by MLN4924. We here show that activation of the ORF50 gene by MLN4924 involves the relief of LANA-mediated transcriptional repression. Importantly, we find that LANA is a neddylated protein. To our knowledge, this is the first report showing that neddylation occurs in viral proteins. Additionally, we provide evidence that different concentrations of MLN4924 have opposite effects on TPA-mediated or SB-mediated KSHV lytic cycle activation. Therefore, in clinical application, we propose that MLN4924 needs to be used with caution in combination therapy to treat KSHV-positive subjects.


Asunto(s)
Ciclopentanos/farmacología , Herpesvirus Humano 8/patogenicidad , Proteínas Inmediatas-Precoces/genética , Pirimidinas/farmacología , Sarcoma de Kaposi/patología , Transactivadores/genética , Enzimas Activadoras de Ubiquitina/antagonistas & inhibidores , Activación Viral/efectos de los fármacos , Antígenos Virales/metabolismo , Ácido Butírico/farmacología , Línea Celular Tumoral , Proliferación Celular , Células HEK293 , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas/efectos de los fármacos , Sarcoma de Kaposi/virología , Acetato de Tetradecanoilforbol/farmacología
12.
PLoS Pathog ; 12(10): e1005918, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27698494

RESUMEN

The switch between latency and the lytic cycle of Kaposi's sarcoma-associated herpesvirus (KSHV) is controlled by the expression of virally encoded ORF50 protein. Thus far, the regulatory mechanism underlying the protein stability of ORF50 is unknown. Our earlier studies have demonstrated that a protein abundance regulatory signal (PARS) at the ORF50 C-terminal region modulates its protein abundance. The PARS region consists of PARS-I (aa 490-535) and PARS-II (aa 590-650), and mutations in either component result in abundant expression of ORF50. Here, we show that ORF50 protein is polyubiquitinated and its abundance is controlled through the proteasomal degradation pathway. The PARS-I motif mainly functions as a nuclear localization signal in the control of ORF50 abundance, whereas the PARS-II motif is required for the binding of ubiquitin enzymes in the nucleus. We find that human oncoprotein MDM2, an ubiquitin E3 ligase, is capable of interacting with ORF50 and promoting ORF50 degradation in cells. The interaction domains between both proteins are mapped to the PARS region of ORF50 and the N-terminal 220-aa region of MDM2. Additionally, we identify lysine residues at positions 152 and 154 in the N-terminal domain of ORF50 critically involved in MDM2-mediated downregulation of ORF50 levels. Within KSHV-infected cells, the levels of MDM2 were greatly reduced during viral lytic cycle and genetic knockdown of MDM2 in these cells favored the enhancement of ORF50 expression, supporting that MDM2 is a negative regulator of ORF50 expression. Collectively, the study elucidates the regulatory mechanism of ORF50 stability and implicates that MDM2 may have a significant role in the maintenance of viral latency by lowering basal level of ORF50.


Asunto(s)
Regulación Viral de la Expresión Génica/fisiología , Infecciones por Herpesviridae/metabolismo , Proteínas Inmediatas-Precoces/biosíntesis , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Transactivadores/biosíntesis , Latencia del Virus/fisiología , Línea Celular , Técnica del Anticuerpo Fluorescente , Herpesvirus Humano 8 , Humanos , Immunoblotting , Inmunoprecipitación , Microscopía Confocal , Estabilidad Proteica
13.
Medicine (Baltimore) ; 95(35): e4701, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27583901

RESUMEN

Ingestion of a foreign body is common among children. However, ingestion of foam earplugs (FEPs) has not been reported previously. A 7-month-old female infant presented with small bowel obstruction, which was finally proved to be a case of FEP ingestion.Computed tomography (CT) phantom study was performed to examine the imaging features of FEPs. We studied the following dry and fully wet FEPs, FEPs squeezed in pure water to varying degrees, and FEPs with different degrees of compression in the dry and wet states from day 0 to 6 and all scanned with a CT scanner.The density of a dry FEP is -843.5 ±â€Š4.5 Hounsfield units (HU) and it increases to 0.76 ±â€Š9.3 HU when fully wet. The densities of FEPs ranged from -844.2 to 1.0 HU with different water/air ratios, and some showed a heterogeneous geographic pattern. The densities of FEPs increase due to compression and gradual water absorption.FEPs can be potentially hazardous objects to children. Owing to the special foam structure of the FEP, it can mimic a fatty lesion if the density ranges from -100 to -50 HU; moreover, it can hide in the water if fully wet. However, it should not be mistaken as air, as the density of a dry FEP is -843.5 HU, and the contour can be observed if the window level is set appropriately. Because of its soft texture, the surgeon should be careful not to miss an FEP during the operation. Moreover, radiologists should be familiar with the CT features of FEPs so that they can be identified before surgery.


Asunto(s)
Dispositivos de Protección de los Oídos , Cuerpos Extraños/diagnóstico por imagen , Íleon/diagnóstico por imagen , Fantasmas de Imagen , Tomografía Computarizada por Rayos X/métodos , Femenino , Cuerpos Extraños/complicaciones , Humanos , Enfermedades del Íleon/etiología , Enfermedades del Íleon/cirugía , Íleon/cirugía , Lactante , Obstrucción Intestinal/etiología , Obstrucción Intestinal/cirugía
14.
J Virol ; 88(17): 10092-109, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24965448

RESUMEN

UNLABELLED: The orf47-orf46-orf45 gene cluster of Kaposi's sarcoma-associated herpesvirus (KSHV) is known to serially encode glycoprotein L (gL), uracil DNA glycosylase, and a viral tegument protein. Here, we identify two novel mRNA variants, orf47/45-A and orf47/45-B, alternatively spliced from a tricistronic orf47-orf46-orf45 mRNA that is expressed in the orf47-orf46-orf45 gene locus during the early stages of viral reactivation. The spliced gene products, ORF47/45-A and ORF47/45-B, consist of only a partial region of gL (ORF47), a unique 7-amino-acid motif, and the complete tegument protein ORF45. Like the ORF45 protein, ORF47/45-A and ORF47/45-B expressed in cells sufficiently activate the phosphorylation of p90 ribosomal S6 kinase (RSK) and extracellular signal-regulated protein kinase (ERK). However, unlike ORF45, both ORF47/45-A and ORF47/45-B contain a signal peptide sequence and are localized at the endoplasmic reticulum (ER). Additionally, we found that ORF47/45-A and ORF47/45-B have an extra function that mediates the upregulation of GRP78, a master regulator of ER homeostasis. The important event regarding GRP78 upregulation can be observed in all tested KSHV-positive cell lines after viral reactivation, and knockdown of GRP78 in cells significantly impairs viral lytic cycle progression, especially at late lytic stages. Compared with some other viral glycoproteins synthesized through the ER, our results strongly implicate that the ORF47/45 proteins may serve as key effectors for controlling GRP78 expression and ER homeostasis in cells. Taken together, our findings provide evidence showing the reciprocal association between the modulation of ER homeostasis and the progression of the KSHV lytic cycle. IMPORTANCE: Emerging evidence has shown that several viruses appear to use different strategies to control ER homeostasis for supporting their productive infections. The two parts of this study identify two aspects of the association between the regulation of ER homeostasis and the progression of the KSHV lytic cycle. The first part characterizes the function of two early lytic cycle proteins, ORF47/45-A and ORF47/45-B, on the activation of a major ER chaperone protein, GRP78. In addition to the ability to promote GRP78 upregulation, the ORF47/45 proteins also activate the phosphorylation of RSK and ERK. The second part reveals that upregulation of GRP78 is essential for the progression of the KSHV lytic cycle, especially at late stages. We therefore propose that activation of GRP78 expression by viral proteins at the early lytic stage may aid with the protection of host cells from severe ER stress and may directly involve the assembly or release of virions.


Asunto(s)
ADN Recombinante , Genes Virales , Herpesvirus Humano 8/genética , Proteínas Inmediatas-Precoces/genética , Familia de Multigenes , ADN Viral/química , ADN Viral/genética , Chaperón BiP del Retículo Endoplásmico , Regulación Viral de la Expresión Génica , Herpesvirus Humano 8/fisiología , Humanos , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Viral/genética , Análisis de Secuencia de ADN , Replicación Viral
15.
Virology ; 442(1): 38-50, 2013 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-23601787

RESUMEN

The ORF45 gene of Kaposi's sarcoma-associated herpesvirus (KSHV) encodes a multifunctional tegument protein. Here, we characterize the transcriptional control of the ORF45 gene and show that its promoter can be activated by ORF50 protein, a latent-lytic switch transactivator. The ORF45 promoter can also be induced by sodium butyrate (SB), a histone deacetylase inhibitor, in the absence of ORF50 protein. Although SB induces the ORF45 gene independently of ORF50, its full activation may require the presence of ORF50. Deletion and point mutation analyses revealed that two RBP-Jκ-binding sites in the ORF45 promoter confer the ORF50 responsiveness, whereas NF-Y and Sp1-binding sites mediate the response to SB. Direct binding of NF-Y, Sp1, or RBP-Jκ protein to the ORF45 promoter is required for the promoter activation induced by SB or by ORF50. In conclusion, our study demonstrates both ORF50-dependent and ORF50-independent transcriptional mechanisms operated on the activation of the ORF45 gene.


Asunto(s)
Regulación Viral de la Expresión Génica , Herpesvirus Humano 8/fisiología , Proteínas Inmediatas-Precoces/metabolismo , Transactivadores/metabolismo , Activación Viral , Sitios de Unión , Butiratos/metabolismo , Butiratos/farmacología , Factor de Unión a CCAAT , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Proteínas Inmediatas-Precoces/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Proteínas Quinasas , Transactivadores/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo
16.
Virus Res ; 165(2): 157-69, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22366521

RESUMEN

The ORF46 gene of Kaposi's sarcoma-associated herpesvirus (KSHV) encodes uracil DNA glycosylase, an enzyme involved in DNA repair. In this study, we show that the transcriptional start site of the ORF46 gene is located at nucleotide 69,425 of the viral genome and ORF50 protein, a latent-lytic switch transactivator, activates the ORF46 promoter via RBP-Jκ protein. Three consensus RBP-Jκ-binding sites found in the ORF46 promoter are critical for the binding of RBP-Jκ protein and conferring the ORF50 responsiveness. In addition, a negative regulatory region has been determined in the ORF46 promoter, which mediates the suppression of the ORF50 responsiveness. The functional negative region of the ORF46 promoter is mainly composed of the Sp1-binding sites. Like the negative region of the ORF46 promoter, addition of Sp1-binding sequences alone in an ORF50-responsive promoter efficiently confers the suppression of the ORF50 responsiveness. Furthermore, sodium butyrate, a pleiotropic inducing agent for the KSHV lytic cycle, is able to relieve the negative regulation of the ORF46 promoter in the latently KSHV-infected cells. The identification of multiple positive and negative cis-acting regulatory elements in the viral promoters emphasizes the elaborate controls in the KSHV lytic cycle, which ensure the adequate expression of each viral lytic gene.


Asunto(s)
Regulación Viral de la Expresión Génica , Herpesvirus Humano 8/genética , Proteínas Inmediatas-Precoces/metabolismo , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Regiones Promotoras Genéticas , Transactivadores/metabolismo , Uracil-ADN Glicosidasa/biosíntesis , Sitios de Unión , Línea Celular Tumoral , Herpesvirus Humano 8/enzimología , Humanos , Unión Proteica , Factor de Transcripción Sp1/metabolismo , Sitio de Iniciación de la Transcripción
17.
Virology ; 413(2): 194-204, 2011 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-21376359

RESUMEN

Lytic cycle reactivation of Kaposi's sarcoma-associated herpesvirus (KSHV) is initiated by expression of the ORF50 gene. Here we show that YY1 protein specifically binds to the ORF50 promoter (ORF50p) region in vitro and in vivo. After treatment with chemical inducers, including sodium butyrate (SB) and TPA, the levels of YY1 protein are inversely correlated with the lytic induction of KSHV in cells. Overexpression of YY1 completely blocks the ORF50p activation in transient reporter assays, while mutation at the YY1 site in the ORF50p or knockdown of YY1 protein confers an enhancement of the ORF50p activation induced by SB and TPA. YY1 overexpression in a stable cell clone HH-B2(Dox-YY1) also inhibits expression of the ORF50 and its downstream lytic genes. On the other hand, a chimeric YY1 construct that links to its coactivator E1A can disrupt viral latency. These results imply that YY1 is involved in the regulation of KSHV reactivation.


Asunto(s)
Herpesvirus Humano 8/fisiología , Proteínas Inmediatas-Precoces/metabolismo , Transactivadores/metabolismo , Factor de Transcripción YY1/metabolismo , Línea Celular , Regulación hacia Abajo , Regulación Viral de la Expresión Génica/fisiología , Humanos , Proteínas Inmediatas-Precoces/genética , Regiones Promotoras Genéticas , Unión Proteica , Interferencia de ARN , Transactivadores/genética , Latencia del Virus , Factor de Transcripción YY1/genética
18.
Pediatr Neonatol ; 51(3): 186-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20675245

RESUMEN

Colonic atresia is a very rare cause of intestinal obstruction, and surgical management is the mainstay of therapy. A case of congenital colonic atresia is reported in a full-term neonate who presented with delayed passage of meconium, abdominal distention and bilious vomiting. The present case and the pertinent literature are discussed, with an emphasis on surgical management.


Asunto(s)
Enfermedades del Colon/congénito , Enfermedades del Colon/cirugía , Atresia Intestinal/cirugía , Obstrucción Intestinal/congénito , Obstrucción Intestinal/cirugía , Sulfato de Bario , Enfermedades del Colon/diagnóstico por imagen , Medios de Contraste , Enema , Femenino , Humanos , Recién Nacido , Atresia Intestinal/diagnóstico por imagen , Obstrucción Intestinal/diagnóstico por imagen , Radiografía
19.
Virology ; 397(2): 311-21, 2010 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-19969319

RESUMEN

The ORF61 and ORF60 genes of Kaposi's sarcoma-associated herpesvirus (KSHV) encode the ribonucleotide reductase large and small subunits, respectively. Here we show that ORF50 protein, a latent-lytic switch transactivator, activates the transcription of these two early-lytic genes through different mechanisms. Activation of the ORF61 promoter by ORF50 protein is dependent on an intact RBP-Jkappa-binding site within the identified responsive element and the expression of RBP-Jkappa protein in cells. The critical element in the ORF60 promoter in response to ORF50 was mapped to a 40-bp region. Binding of YY1, Sp1/Sp3 or unknown proteins to this element may contribute to repression or activation of the ORF60 promoter. Although ORF50 protein does not directly bind to the ORF61 and ORF60 promoters in vitro, we show the association of ORF50 protein with these two promoters in vivo. Our results provide further insights into the regulatory network of the viral lytic genes in KSHV reactivation.


Asunto(s)
Regulación Viral de la Expresión Génica , Herpesvirus Humano 8/fisiología , Sistemas de Lectura Abierta , Transcripción Genética , Proteínas Virales/fisiología , Sitios de Unión , ADN Viral/metabolismo , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Regiones Promotoras Genéticas , Unión Proteica
20.
Kaohsiung J Med Sci ; 23(9): 435-41, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17766211

RESUMEN

In patients who require a permanent central venous catheter (PCVC), the usual aim is to put the catheter tip at the superior vena cava and right atrium (SVC-RA) junction. However, there is no study regarding how to guide the positioning of the catheter tip when the SVC-RA junction cannot be identified on chest radiograph. The objectives of this prospective study were: (1) to investigate the incidence and etiologies of radiographically undetermined SVC-RA junctions in cancer patients undergoing PCVC implantation; and (2) to evaluate the feasibility, effectiveness and safety of combined transesophageal echocardiography (TEE) and laryngeal mask airway (LMA) to guide the positioning of catheters during implantations in patients without this radiographic landmark. Over a 1-year study period, 83 consecutive patients with oncologic diseases who required implantation of a PCVC in a tertiary center were screened. Their preoperative chest radiographs were examined by radiologists to identify the presence of the SVC-RA junction. Patients without a radiographically identifiable SVC-RA junction were classified as cancer-related or cancer-unrelated in terms of etiology. For patients without this landmark, we used TEE with a pediatric biplane transducer and a LMA under intravenous general anesthesia during PCVC implantation to guide the positioning of the catheter tip at the SVC-RA junction. We found that in 16% (13/83) of patients, the SVC-RA junction could not be identified on radiograph. Among the 13 patients, only three (23%) had cancer-related etiologies. In all of the 13 patients, the LMA was successfully placed after the TEE transducer was inserted. No episode of air leak from the LMA was found during surgery. All had the catheter tip positioned in the anatomic SVC-RA junction under TEE guidance. In conclusion, 16% of cancer patients requiring PCVC implantation had no identifiable SVC-RA junction on chest radiograph and most causes were cancer-unrelated. In patients without a radiographically identifiable SVC-RA junction, guidance by TEE under LMA general anesthesia is a feasible, safe and effective management to position a PCVC at the SVC-RA junction.


Asunto(s)
Cateterismo Venoso Central/métodos , Ecocardiografía Transesofágica , Atrios Cardíacos/diagnóstico por imagen , Máscaras Laríngeas , Neoplasias/terapia , Vena Cava Superior/diagnóstico por imagen , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Radiografía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA