Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Dialogues Health ; 2: 100117, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38515494

RESUMO

Background: This work aims to analyze the landscape of scientific publications on subjects related to One Health and infectious diseases in Panama. The research questions are: How does the One Health research landscape look like in Panama? Are historical research efforts aligned with the One Health concept? What infectious diseases have received more attention from the local scientific community since 1990? Methods: Boolean searches on the Web of Science, SCOPUS and PubMed were undertaken to evaluate the main trends of publications related to One Health and infectious disease research in the country of Panama, between 1990 and 2019. Results: 4546 publications were identified since 1990, including 3564 peer-reviewed articles interconnected with One Health related descriptors, and 211 articles focused particularly on infectious diseases. A pattern of exponential growth in the number of publications with various contributions from Panamanian institutions was observed. The rate of multidisciplinary research was moderate, whereas those of interinstitutional and intersectoral research ranged from low to very low. Research efforts have centered largely on protozoan, neglected and arthropod-borne diseases with a strong emphasis on malaria, Chagas and leishmaniasis. Conclusion: Panama has scientific capabilities on One Health to tackle future infectious disease threats, but the official collaboration schemes and strategic investment to develop further competencies need to be conciliated with modern times, aka the pandemics era. The main proposition here, addressed to the government of Panama, is to launch a One Health regional center to promote multidisciplinary, interinstitutional and intersectoral research activities in Panama and beyond.

2.
Molecules ; 27(21)2022 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-36364188

RESUMO

Mayaro virus (MAYV) is an emerging arbovirus with an increasing circulation across the Americas. In the present study, we evaluated the potential antiviral activity of the following natural compounds against MAYV and other arboviruses: Sanguinarine, (R)-Shikonin, Fisetin, Honokiol, Tanshinone IIA, and α-Mangostin. Sanguinarine and Shikonin showed significant cytotoxicity, whereas Fisetin, Honokiol, Tanshinone IIA, and α-Mangostin were well tolerated in all the cell lines tested. Honokiol and α-Mangostin treatment protected Vero-E6 cells against MAYV-induced damage and resulted in a dose-dependent reduction in viral progeny yields for each of the MAYV strains and human cell lines assessed. These compounds also reduced MAYV viral RNA replication in HeLa cells. In addition, Honokiol and α-Mangostin disrupted MAYV infection at different stages of the virus life cycle. Moreover, Honokiol and α-Mangostin decreased Una, Chikungunya, and Zika viral titers and downmodulated the expression of E1 and nsP1 viral proteins from MAYV, Una, and Chikungunya. Finally, in Honokiol- and α-Mangostin-treated HeLa cells, we observed an upregulation in the expression of type I interferon and specific interferon-stimulated genes, including IFNα, IFNß, MxA, ISG15, OAS2, MDA-5, TNFα, and IL-1ß, which may promote an antiviral cellular state. Our results indicate that Honokiol and α-Mangostin present potential broad-spectrum activity against different arboviruses through different mechanisms.


Assuntos
Alphavirus , Arbovírus , Febre de Chikungunya , Infecção por Zika virus , Zika virus , Humanos , Células HeLa , Alphavirus/genética , Replicação Viral , Antivirais/farmacologia
4.
Front Cell Infect Microbiol ; 12: 960138, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35967877

RESUMO

Active hypusine-modified initiation elongation factor 5A is critical for cell proliferation and differentiation, embryonic development, and innate immune response of macrophages to bacterial infection. Here, we demonstrate that both virus infection and double-stranded RNA viral mimic stimulation induce the hypusination of eIF5A. Furthermore, we show that activation of eIF5A is essential for the replication of several RNA viruses including influenza A virus, vesicular stomatitis virus, chikungunya virus, mayaro virus, una virus, zika virus, and punta toro virus. Finally, our data reveal that inhibition of eIF5A hypusination using the spermidine analog GC7 or siRNA-mediated downmodulation of eIF5A1 induce upregulation of endoplasmic reticulum stress marker proteins and trigger the transcriptional induction of interferon and interferon-stimulated genes, mechanisms that may explain the broad-spectrum antiviral activity of eIF5A inhibition.


Assuntos
Vírus de RNA , Viroses , Infecção por Zika virus , Zika virus , Antivirais , Humanos , Interferons , RNA de Cadeia Dupla , Replicação Viral
5.
Viruses ; 14(2)2022 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-35216015

RESUMO

Mayaro virus (MAYV) manipulates cell machinery to successfully replicate. Thus, identifying host proteins implicated in MAYV replication represents an opportunity to discover potential antiviral targets. PIM kinases are enzymes that regulate essential cell functions and also appear to be critical factors in the replication of certain viruses. In this study we explored the consequences of PIM kinase inhibition in the replication of MAYV and other arboviruses. Cytopathic effects or viral titers in samples from MAYV-, Chikungunya-, Una- or Zika-infected cells treated with PIM kinase inhibitors were evaluated using an inverted microscope or plaque-forming assays. The expression of viral proteins E1 and nsP1 in MAYV-infected cells was assessed using an immunofluorescence confocal microscope or Western blot. Our results revealed that PIM kinase inhibition partially prevented MAYV-induced cell damage and also promoted a decrease in viral titers for MAYV, UNAV and ZIKV. The inhibitory effect of PIM kinase blocking was observed for each of the MAYV strains tested and also occurred as late as 8 h post infection (hpi). Finally, PIM kinase inhibition suppressed the expression of MAYV E1 and nsP1 proteins. Taken together, these findings suggest that PIM kinases could represent an antiviral target for MAYV and other arboviruses.


Assuntos
Alphavirus/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-pim-1/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos , Animais , Antivirais/farmacologia , Linhagem Celular , Vírus Chikungunya/efeitos dos fármacos , Humanos , Zika virus/efeitos dos fármacos
6.
Viruses ; 13(6)2021 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-34204188

RESUMO

Mayaro virus (MAYV) hijacks the host's cell machinery to effectively replicate. The mitogen-activated protein kinases (MAPKs) p38, JNK, and ERK1/2 have emerged as crucial cellular factors implicated in different stages of the viral cycle. However, whether MAYV uses these MAPKs to competently replicate has not yet been determined. The aim of this study was to evaluate the impact of MAPK inhibition on MAYV replication using primary human dermal fibroblasts (HDFs) and HeLa cells. Viral yields in supernatants from MAYV-infected cells treated or untreated with inhibitors SB203580, SP600125, U0126, or Losmapimod were quantified using plaque assay. Additionally, viral protein expression was analyzed using immunoblot and immunofluorescence. Knockdown of p38⍺/p38ß isoforms was performed in HDFs using the PROTACs molecule NR-7h. Our data demonstrated that HDFs are highly susceptible to MAYV infection. SB203580, a p38 inhibitor, reduced MAYV replication in a dose-dependent manner in both HDFs and HeLa cells. Additionally, SB203580 significantly decreased viral E1 protein expression. Similarly, knockdown or inhibition of p38⍺/p38ß isoforms with NR-7h or Losmapimod, respectively, affected MAYV replication in a dose-dependent manner. Collectively, these findings suggest that p38 could play an important role in MAYV replication and could serve as a therapeutic target to control MAYV infection.


Assuntos
Alphavirus/fisiologia , Fibroblastos/virologia , Replicação Viral/genética , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Apoptose , Células Cultivadas , Ciclopropanos/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Células HeLa , Interações entre Hospedeiro e Microrganismos/efeitos dos fármacos , Humanos , Imidazóis/farmacologia , Sistema de Sinalização das MAP Quinases , Fosforilação , Piridinas/farmacologia , Pele/citologia , Pele/virologia , Replicação Viral/efeitos dos fármacos
7.
Viruses ; 12(4)2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32326564

RESUMO

The alphaviruses Chikungunya (CHIKV), Mayaro (MAYV), Una (UNAV), and the flavivirus Zika (ZIKV) are emerging or re-emerging arboviruses which are responsible for frequent epidemic outbreaks. Despite the large impact of these arboviruses on health systems, there are no approved vaccines or treatments to fight these infections. As a consequence, there is an urgent need to discover new antiviral drugs. Natural products are a rich source of compounds with distinct biological activities, including antiviral properties. Thus, we aimed to explore the potential antiviral activity of Ginkgolic acid against the arboviruses CHIKV, MAYV, UNAV, and ZIKV. Viral progeny production in supernatants from cells treated or not treated with Ginkgolic acid was quantified by plaque-forming assay. Ginkgolic acid's direct virucidal activity against these arboviruses was also determined. Additionally, viral protein expression was assessed using Western blot and immunofluorescence. Our results reveal that Ginkgolic acid promotes a dose-dependent decrease in viral titers in all tested viruses. Moreover, the compound demonstrated strong virucidal activity. Finally, we found that viral protein expression was affected by treatment with this drug. Collectively, these findings suggest that Ginkgolic acid could have broader antiviral activity.


Assuntos
Alphavirus/efeitos dos fármacos , Antivirais/farmacologia , Vírus Chikungunya/efeitos dos fármacos , Salicilatos/farmacologia , Zika virus/efeitos dos fármacos , Animais , Células Cultivadas , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Células Vero , Ensaio de Placa Viral , Replicação Viral/efeitos dos fármacos
8.
Adv Exp Med Biol ; 1233: 263-277, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32274761

RESUMO

SUMO is a ubiquitin-like protein that covalently binds to lysine residues of target proteins and regulates many biological processes such as protein subcellular localization or stability, transcription, DNA repair, innate immunity, or antiviral defense. SUMO has a critical role in the signaling pathway governing type I interferon (IFN) production, and among the SUMOylation substrates are many IFN-induced proteins. The overall effect of IFN is increasing global SUMOylation, pointing to SUMO as part of the antiviral stress response. Viral agents have developed different mechanisms to counteract the antiviral activities exerted by SUMO, and some viruses have evolved to exploit the host SUMOylation machinery to modify their own proteins. The exploitation of SUMO has been mainly linked to nuclear replicating viruses due to the predominant nuclear localization of SUMO proteins and enzymes involved in SUMOylation. However, SUMOylation of numerous viral proteins encoded by RNA viruses replicating at the cytoplasm has been lately described. Whether nuclear localization of these viral proteins is required for their SUMOylation is unclear. Here, we summarize the studies on exploitation of SUMOylation by cytoplasmic RNA viruses and discuss about the requirement for nuclear localization of their proteins.


Assuntos
Citoplasma/virologia , Vírus de RNA/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Proteínas Virais/metabolismo , Núcleo Celular/metabolismo , Humanos , Sumoilação
9.
Viruses ; 11(4)2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-31018496

RESUMO

Mayaro (MAYV) and Una (UNAV) are emerging arboviruses belonging to the Alphavirus genus of the Togaviridae family. These viruses can produce febrile disease with symptoms such as fever, headache, myalgia, skin rash and incapacitating poly-arthralgia. Serological studies indicate that both viruses are circulating in different countries in Latin America. Viruses need the host cell machinery and resources to replicate effectively. One strategy to find new antivirals consists of identifying key cellular pathways or factors that are essential for virus replication. In this study, we analyzed the role of the ubiquitin-proteasome system (UPS) in MAYV and UNAV replication. Vero-E6 or HeLa cells were treated with the proteasome inhibitors MG132 or Lactacystin, and viral progeny production was quantified using a plaque assay method. In addition, the synthesis of viral proteins was analyzed by Western blot and confocal microscopy. Our results indicate that treatment with proteasome inhibitors decreases MAYV and UNAV protein synthesis, and also causes a significant dose-dependent decrease in MAYV and UNAV replication. Proteasome activity seems to be important at the early stages of MAYV replication. These findings suggest that the ubiquitin-proteasome system is a possible pharmacological target to inhibit these neglected alphaviruses.


Assuntos
Alphavirus/efeitos dos fármacos , Antivirais/farmacologia , Complexo de Endopeptidases do Proteassoma/fisiologia , Replicação Viral , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Alphavirus/fisiologia , Animais , Chlorocebus aethiops , Inibidores de Cisteína Proteinase/farmacologia , Citoplasma/efeitos dos fármacos , Citoplasma/virologia , Células HeLa , Humanos , Leupeptinas/farmacologia , Inibidores de Proteassoma/farmacologia , Células Vero
10.
Cardiovasc Res ; 109(1): 67-78, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26260798

RESUMO

AIMS: After myocardial infarction (MI), extensive remodelling of the extracellular matrix contributes to scar formation. While aiming to preserve tissue integrity, this fibrotic response is also associated with adverse events, including a markedly increased risk of heart failure, ventricular arrhythmias, and sudden cardiac death. Cardiac fibrosis is characterized by extensive deposition of collagen and also by increased stiffness as a consequence of enhanced collagen cross-linking. Members of the lysyl oxidase (LOX) family of enzymes are responsible for the formation of collagen cross-links. This study investigates the contribution of LOX family members to the heart response to MI. METHODS AND RESULTS: Experimental MI was induced in C57BL/6 mice by permanent ligation of the left anterior descending coronary artery. The expression of LOX isoforms (LOX and LOXL1-4) was strongly increased upon MI, and this response was accompanied by a significant accumulation of mature collagen fibres in the infarcted area. LOX expression was observed in areas of extensive remodelling, partially overlapping with α-smooth muscle actin-expressing myofibroblasts. Tumour growth factor-ß as well as hypoxia-activated pathways contributed to the induction of LOX expression in cardiac fibroblasts. Finally, in vivo post-infarction treatment with the broadband LOX inhibitor ß-aminopropionitrile or, selectively, with a neutralizing antibody against the canonical LOX isoform attenuated collagen accumulation and maturation and also resulted in reduced ventricular dilatation and improved cardiac function. CONCLUSION: LOX family members contribute significantly to the detrimental effects of cardiac remodelling, highlighting LOX inhibition as a potential therapeutic strategy for post-infarction recovery.


Assuntos
Matriz Extracelular/fisiologia , Coração/fisiopatologia , Infarto do Miocárdio/enzimologia , Proteína-Lisina 6-Oxidase/biossíntese , Animais , Hipóxia Celular , Células Cultivadas , Indução Enzimática , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/fisiopatologia , Proteína-Lisina 6-Oxidase/antagonistas & inibidores , Proteína-Lisina 6-Oxidase/genética , Fator de Crescimento Transformador beta/farmacologia
11.
Life Sci ; 118(2): 156-64, 2014 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-24378671

RESUMO

The endothelin (ET) system consists of two G-protein-coupled receptors (ETA and ETB), three peptide ligands (ET-1, ET-2 and ET-3), and two activating peptidases (endothelin-converting enzyme-, ECE-1 and ECE-2). While initially described as a vasoregulatory factor, shown to influence several cardiovascular diseases, from hypertension to heart failure, ET-1, the predominant form in most cells and tissues, has expanded its pathophysiological relevance by recent evidences implicating this factor in the regulation of fibrosis. In this article, we review the current knowledge of the role of ET-1 in the development of fibrosis, with particular focus on the regulation of its biosynthesis and the molecular mechanisms involved in its profibrotic actions. We summarize also the contribution of ET-1 to fibrotic disorders in several organs and tissues. The development and availability of specific ET receptor antagonists have greatly stimulated a number of clinical trials in these pathologies that unfortunately have so far given negative or inconclusive results. This review finally discusses the circumstances underlying these disappointing results, as well as provides basic and clinical researchers with arguments to keep exploring the complex physiology of ET-1 and its therapeutic potential in the process of fibrosis.


Assuntos
Endotelina-1/metabolismo , Fibrose/terapia , Animais , Endotelina-1/genética , Fibrose/genética , Humanos , Modelos Biológicos , Especificidade de Órgãos
12.
Mol Cell Biol ; 33(12): 2388-401, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23572561

RESUMO

Transforming growth factor ß1 (TGF-ß1) is a pleiotropic factor involved in the regulation of extracellular matrix (ECM) synthesis and remodeling. In search for novel genes mediating the action of TGF-ß1 on vascular ECM, we identified the member of the lysyl oxidase family of matrix-remodeling enzymes, lysyl oxidase-like 4 (LOXL4), as a direct target of TGF-ß1 in aortic endothelial cells, and we dissected the molecular mechanism of its induction. Deletion mapping and mutagenesis analysis of the LOXL4 promoter demonstrated the absolute requirement of a distal enhancer containing an activator protein 1 (AP-1) site and a Smad binding element for TGF-ß1 to induce LOXL4 expression. Functional cooperation between Smad proteins and the AP-1 complex composed of JunB/Fra2 accounted for the action of TGF-ß1, which involved the extracellular signal-regulated kinase (ERK)-dependent phosphorylation of Fra2. We furthermore provide evidence that LOXL4 was extracellularly secreted and significantly contributed to ECM deposition and assembly. These results suggest that TGF-ß1-dependent expression of LOXL4 plays a role in vascular ECM homeostasis, contributing to vascular processes associated with ECM remodeling and fibrosis.


Assuntos
Antígeno 2 Relacionado a Fos/metabolismo , Proteína-Lisina 6-Oxidase/metabolismo , Proteínas Smad/metabolismo , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Aorta/metabolismo , Sequência de Bases , Sítios de Ligação/genética , Sítios de Ligação/fisiologia , Bovinos , Células Cultivadas , Células Endoteliais/metabolismo , Matriz Extracelular/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Antígeno 2 Relacionado a Fos/genética , Células HEK293 , Humanos , Camundongos , Fosforilação , Regiões Promotoras Genéticas , Proteína-Lisina 6-Oxidase/biossíntese , Proteína-Lisina 6-Oxidase/genética , Interferência de RNA , RNA Interferente Pequeno , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Fator de Transcrição AP-1/metabolismo , Fatores de Transcrição/genética
13.
J Virol ; 87(2): 807-17, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23115286

RESUMO

Posttranslational modification by SUMO provides functional flexibility to target proteins. Viruses interact extensively with the cellular SUMO modification system in order to improve their replication, and there are numerous examples of viral proteins that are SUMOylated. However, thus far the relevance of SUMOylation for rotavirus replication remains unexplored. In this study, we report that SUMOylation positively regulates rotavirus replication and viral protein production. We show that SUMO can be covalently conjugated to the viroplasm proteins VP1, VP2, NSP2, VP6, and NSP5. In addition, VP1, VP2, and NSP2 can also interact with SUMO in a noncovalent manner. We observed that an NSP5 SUMOylation mutant protein retains most of its activities, such as its interaction with VP1 and NSP2, the formation of viroplasm-like structures after the coexpression with NSP2, and the ability to complement in trans the lack of NSP5 in infected cells. However, this mutant is characterized by a high degree of phosphorylation and is impaired in the formation of viroplasm-like structures when coexpressed with VP2. These results reveal for the first time a positive role for SUMO modification in rotavirus replication, describe the SUMOylation of several viroplasm resident rotavirus proteins, and demonstrate a requirement for NSP5 SUMOylation in the production of viroplasm-like structures.


Assuntos
Interações Hospedeiro-Patógeno , Rotavirus/patogenicidade , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação , Proteínas Virais/metabolismo , Replicação Viral , Animais , Linhagem Celular , Humanos , Ligação Proteica
14.
Cell Cycle ; 10(21): 3701-5, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22033337

RESUMO

Tumor suppressor p53 is known to be a direct transcriptional target of type I interferons (IFNs), contributing to virus-induced apoptosis, and in turn activating itself the interferon pathway. Acetylation, among many other post-translational modifications of p53, is thought to exert a crucial role regulating p53 activity. Here, we examined the contribution of this modification on the antiviral activity mediated by p53. Our results show that virus infection induces p53 acetylation at lysine 379, and that this modification is absolutely required for p53-dependent transcriptional transactivation of both, pro-apoptotic and IFN-stimulated genes induced by virus infection, and for p53-mediated control of virus replication. Thus, our study identifies p53 acetylation as an indispensable event that enables the p53-mediated antiviral response.


Assuntos
Proteína Supressora de Tumor p53/fisiologia , Acetilação , Animais , Antivirais/metabolismo , Apoptose/genética , Regulação da Expressão Gênica , Herpesvirus Humano 1/imunologia , Interferon Tipo I/metabolismo , Lisina/metabolismo , Camundongos , Ativação Transcricional , Proteína Supressora de Tumor p53/metabolismo , Vesiculovirus/imunologia , Replicação Viral
15.
J Virol ; 85(24): 12890-900, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21957283

RESUMO

The vaccinia virus (VACV) E3 protein is essential for virulence and has antiapoptotic activity and the ability to impair the host innate immune response. Here we demonstrate that E3 interacts with SUMO1 through a small ubiquitin-like modifier (SUMO)-interacting motif (SIM). SIM integrity is required for maintaining the stability of the viral protein and for the covalent conjugation of E3 to SUMO1 or SUMO2, a modification that has a negative effect on the E3 transcriptional transactivation of the p53-upregulated modulator of apoptosis (PUMA) and APAF-1 genes. We also demonstrate that E3 is ubiquitinated, a modification that does not destabilize the wild-type protein but triggers the degradation of an E3-ΔSIM mutant. This report constitutes the first demonstration of the important roles that both SUMO and ubiquitin play in the regulation of the VACV protein E3.


Assuntos
Regulação para Baixo , Interações Hospedeiro-Patógeno , Proteínas de Ligação a RNA/metabolismo , Proteína SUMO-1/metabolismo , Vaccinia virus/imunologia , Proteínas Virais/metabolismo , Fatores de Virulência/metabolismo , Linhagem Celular , Humanos , Ligação Proteica , Mapeamento de Interação de Proteínas , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Ubiquitinação
16.
J Gen Virol ; 92(Pt 1): 188-94, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20881090

RESUMO

The multifunctional Kaposi's sarcoma-associated herpesvirus (KSHV) latent protein latency-associated nuclear antigen 2 (LANA2) has a critical role in KSHV-induced B-cell malignancies. LANA2 increases the level of small ubiquitin-like modifier (SUMO)2-ubiquitin-modified PML and induces the disruption of PML oncogenic domains (PODs) by a process that requires a non-covalent SUMO interaction domain (SIM) in LANA2. We now demonstrate that LANA2 is covalently conjugated to SUMO1 and SUMO2 both in vitro and in latently KSHV-infected B-cells. We show that a LANA2 SIM mutant exhibits a slightly altered sumoylation pattern, which suggests that non-covalent SUMO interactions represent a mechanism for determining SUMO substrate recognition and modification. In addition, several lysine residues were mapped as SUMO conjugation sites. A sumoylation-deficient mutant shows impaired ability to induce disruption of PODs, which suggests that either directly bound or covalently conjugated SUMO moieties may act as a bridge for interaction between LANA2 and other SUMO-modified or SUMO-interacting proteins required for disruption of PODs.


Assuntos
Antígenos Virais/metabolismo , Herpesvirus Humano 8/patogenicidade , Proteínas Nucleares/metabolismo , Proteína SUMO-1/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Linfócitos B/virologia , Linhagem Celular , Humanos , Estrutura Terciária de Proteína , Sumoilação , Latência Viral
17.
PLoS One ; 4(7): e6422, 2009 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-19649275

RESUMO

The retinoblastoma protein Rb is a tumor suppressor involved in cell cycle control, differentiation, and inhibition of oncogenic transformation. Besides these roles, additional functions in the control of immune response have been suggested. In the present study we investigated the consequences of loss of Rb in viral infection. Here we show that virus replication is increased by the absence of Rb, and that Rb is required for the activation of the NF-kB pathway in response to virus infection. These results reveal a novel role for tumor suppressor Rb in viral infection surveillance and further extend the concept of a link between tumor suppressors and antiviral activity.


Assuntos
NF-kappa B/metabolismo , Proteína do Retinoblastoma/metabolismo , Estomatite Vesicular/metabolismo , Animais , Células Cultivadas , Camundongos , Camundongos Knockout
18.
J Virol ; 83(17): 8849-58, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19553342

RESUMO

Infection by herpesviruses causes a dramatic disturbance of PML oncogenic domains (PODs) that has been suggested to be essential for viral lytic replication. Several proteins from Kaposi's sarcoma-associated herpesvirus (KSHV) have been tested as putative POD-disrupting factors with negative results. Here, we show that LANA2, a viral protein that is absolutely required for the viability and proliferation of KSHV-infected primary effusion lymphoma (PEL) cells, increases the levels of SUMO2-ubiquitin-modified PML and induces the disruption of PODs by a proteasome-mediated mechanism. In addition, we demonstrate that this disruption is largely dependent on both the integrity of a SUMO interaction motif in LANA2 and the lysine 160 from PML. Moreover, silencing of LANA2 expression in PEL cells by RNA interference led to an increase in the PML levels. Finally, we demonstrate that LANA2 relieves PML-mediated transcriptional repression of survivin, a protein that directly contributes to malignant progression of PEL. This represents the first example of inactivation of these important antiviral structures by KSHV.


Assuntos
Antígenos Virais/metabolismo , Herpesvirus Humano 8/fisiologia , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Proteínas Supressoras de Tumor/antagonistas & inibidores , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes/métodos , Inativação Gênica , Humanos , Proteínas Inibidoras de Apoptose , Proteína da Leucemia Promielocítica , Survivina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...