Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-33495228

RESUMO

Herpesviruses are widespread and can cause serious illness. Many currently available antiviral drugs have limited effects, result in rapid development of resistance, and often exhibit dose-dependent toxicity. Especially for human cytomegalovirus (HCMV), new well-tolerated compounds with novel mechanisms of action are urgently needed. In this study, we characterized the antiviral activity of two new diazadispiroalkane derivatives, 11826091 and 11826236. These two small molecules exhibited strong activity against low-passage-number HCMV. Pretreatment of cell-free virus with these compounds greatly reduced infection. Time-of-addition assays where 11826091 or 11826236 was added to cells before infection, before and during infection, or during or after infection demonstrated an inhibitory effect on early steps of infection. Interestingly, 11826236 had an effect by addition to cells after infection. Results from entry assays showed the major effect to be on attachment. Only 11826236 had a minimal effect on penetration comparable to heparin. Further, no effect on virus infection was found for cell lines with a defect in heparan sulfate expression or lacking all surface glycosaminoglycans, indicating that these small molecules bind to heparan sulfate on the cell surface. To test this further, we extended our analyses to pseudorabies virus (PrV), a member of the Alphaherpesvirinae, which is known to use cell surface heparan sulfate for initial attachment via nonessential glycoprotein C (gC). While infection with PrV wild type was strongly impaired by 11826091 or 11826236, as with heparin, a mutant lacking gC was unaffected by either treatment, demonstrating that primary attachment to heparan sulfate via gC is targeted by these small molecules.


Assuntos
Herpesvirus Suídeo 1 , Internalização do Vírus , Alcanos , Animais , Antivirais , Glicosaminoglicanos , Heparina/farmacologia , Heparitina Sulfato , Humanos , Compostos de Espiro , Proteínas do Envelope Viral
2.
PLoS Pathog ; 15(12): e1008175, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31809525

RESUMO

A key step in replication of human cytomegalovirus (HCMV) in the host cell is the generation and packaging of unit-length genomes into preformed capsids. The enzymes involved in this process are the terminases. The HCMV terminase complex consists of two terminase subunits, the ATPase pUL56 and the nuclease pUL89. A potential third component pUL51 has been proposed. Even though the terminase subunit pUL89 has been shown to be essential for DNA packaging and interaction with pUL56, it is not known how pUL89 mechanistically achieves sequence-specific DNA binding and nicking. To identify essential domains and invariant amino acids vis-a-vis nuclease activity and DNA binding, alanine substitutions of predicted motifs were analyzed. The analyses indicated that aspartate 463 is an invariant amino acid for the nuclease activity, while argine 544 is an invariant aa for DNA binding. Structural analysis of recombinant protein using electron microscopy in conjunction with single particle analysis revealed a curvilinear monomer with two distinct domains connected by a thinner hinge-like region that agrees well with the predicted structure. These results allow us to model how the terminase subunit pUL89's structure may mediate its function.


Assuntos
Citomegalovirus/química , Empacotamento do DNA/fisiologia , Proteínas Virais/química , Citomegalovirus/genética , Conformação Proteica , Relação Estrutura-Atividade , Proteínas Virais/genética
3.
J Virol ; 90(13): 5876-5885, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27053556

RESUMO

UNLABELLED: DNA packaging into procapsids is a common multistep process during viral maturation in herpesviruses. In human cytomegalovirus (HCMV), the proteins involved in this process are terminase subunits pUL56 and pUL89, which are responsible for site-specific cleavage and insertion of the DNA into the procapsid via portal protein pUL104. However, additional viral proteins are required for the DNA packaging process. We have shown previously that the plasmid that encodes capsid-associated pUL77 encodes another potential player during capsid maturation. Pulse-chase experiments revealed that pUL77 is stably expressed during HCMV infection. Time course analysis demonstrated that pUL77 is expressed in the early late part of the infectious cycle. The sequence of pUL77 was analyzed to find nuclear localization sequences (NLSs), revealing monopartite NLSm at the N terminus and bipartite NLSb in the middle of pUL77. The potential NLSs were inserted into plasmid pHM829, which encodes a chimeric protein with ß-galactosidase and green fluorescent protein. In contrast to pUL56, neither NLSm nor NLSb was sufficient for nuclear import. Furthermore, we investigated by coimmunoprecipitation whether packaging proteins, as well as pUL93, the homologue protein of herpes simplex virus 1 pUL17, are interaction partners of pUL77. The interactions between pUL77 and packaging proteins, as well as pUL93, were verified. IMPORTANCE: We showed that the capsid-associated pUL77 is another potential player during capsid maturation of HCMV. Protein UL77 (pUL77) is a conserved core protein of HCMV. This study demonstrates for the first time that pUL77 has early-late expression kinetics during the infectious cycle and an intrinsic potential for nuclear translocation. According to its proposed functions in stabilization of the capsid and anchoring of the encapsidated DNA during packaging, interaction with further DNA packaging proteins is required. We identified physical interactions with terminase subunits pUL56 and pUL89 and another postulated packaging protein, pUL93, in infected, as well as transfected, cells.


Assuntos
Proteínas do Capsídeo/metabolismo , Capsídeo/metabolismo , Citoplasma/química , Empacotamento do DNA , Sinais de Localização Nuclear/química , Proteínas Virais/metabolismo , Capsídeo/química , Citomegalovirus/química , Citomegalovirus/genética , Citomegalovirus/metabolismo , Citoplasma/metabolismo , Citoplasma/virologia , DNA Viral/genética , Endodesoxirribonucleases/metabolismo , Fibroblastos/virologia , Proteínas de Fluorescência Verde/genética , Células HEK293 , Herpesvirus Humano 1/genética , Humanos , Sinais de Localização Nuclear/genética , Sinais de Localização Nuclear/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , beta-Galactosidase/genética
4.
PLoS Pathog ; 10(1): e1003863, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24453968

RESUMO

Nuclear domain 10 (ND10) components are restriction factors that inhibit herpesviral replication. Effector proteins of different herpesviruses can antagonize this restriction by a variety of strategies, including degradation or relocalization of ND10 proteins. We investigated the interplay of Kaposi's Sarcoma-Associated Herpesvirus (KSHV) infection and cellular defense by nuclear domain 10 (ND10) components. Knock-down experiments in primary human cells show that KSHV-infection is restricted by the ND10 components PML and Sp100, but not by ATRX. After KSHV infection, ATRX is efficiently depleted and Daxx is dispersed from ND10, indicating that these two ND10 components can be antagonized by KSHV. We then identified the ORF75 tegument protein of KSHV as the viral factor that induces the disappearance of ATRX and relocalization of Daxx. ORF75 belongs to a viral protein family (viral FGARATs) that has homologous proteins in all gamma-herpesviruses. Isolated expression of ORF75 in primary cells induces a relocalization of PML and dispersal of Sp100, indicating that this viral effector protein is able to influence multiple ND10 components. Moreover, by constructing a KSHV mutant harboring a stop codon at the beginning of ORF75, we could demonstrate that ORF75 is absolutely essential for viral replication and the initiation of viral immediate-early gene expression. Using recombinant viruses either carrying Flag- or YFP-tagged variants of ORF75, we could further corroborate the role of ORF75 in the antagonization of ND10-mediated intrinsic immunity, and show that it is independent of the PML antagonist vIRF3. Members of the viral FGARAT family target different ND10 components, suggesting that the ND10 targets of viral FGARAT proteins have diversified during evolution. We assume that overcoming ND10 intrinsic defense constitutes a critical event in the replication of all herpesviruses; on the other hand, restriction of herpesviral replication by ND10 components may also promote latency as the default outcome of infection.


Assuntos
Infecções por Herpesviridae/imunologia , Herpesvirus Humano 8/fisiologia , Imunidade Inata , Proteínas Nucleares/imunologia , Proteínas Estruturais Virais/imunologia , Replicação Viral/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Células Cultivadas , Proteínas Correpressoras , Códon de Terminação/genética , Códon de Terminação/imunologia , DNA Helicases/genética , DNA Helicases/imunologia , Técnicas de Silenciamento de Genes , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/patologia , Humanos , Masculino , Chaperonas Moleculares , Mutação , Proteínas Nucleares/genética , Proteínas Estruturais Virais/genética , Proteína Nuclear Ligada ao X
5.
Antimicrob Agents Chemother ; 58(4): 1963-71, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24419339

RESUMO

Human cytomegalovirus (HCMV) can cause life-threatening diseases in neonates and immunocompromised patients. Due to multiple problems caused by the current available drugs, development of new antiviral compounds is urgently needed. In this study, we characterize the anti-HCMV spectrum and mechanism of action of the N-N'-(bis-5 nitropyrimidyl)dispirotripiperazine derivate 27 (DSTP-27). DSTP-27 exhibited strong antiviral activity against two laboratory HCMV strains with different cell tropism as well as ganciclovir (GCV)-sensitive and GCV-resistant clinical isolates in plaque reduction assays and viral growth kinetics experiments. Interestingly, neither infectious nor noninfectious viral particles were observed by electron microscopy. Pretreatment of cell-free virus with DSTP-27 prevented virus infection. The results from time of addition assays, in which DTSP-27 was added to cells (i) before infection, (ii) during virus adsorption, or (iii) after adsorption, demonstrated an inhibitory effect on early steps of the HCMV replication cycle. This observation was confirmed by immunofluorescence as well as Western blot analysis, whereby reduced levels of the immediate early protein IE1, the processivity factor pUL44, and the tegument protein pp28 were detected. Results from attachment and penetration analyses of prechilled human embryonic lung fibroblasts revealed that virus attachment is not blocked. In addition, DSTP-27 inactivated HCMV by stable binding. Taken together, these results demonstrate that DSTP-27 (i) blocks viral penetration by interacting with the host cell and (ii) inactivates HCMV by interacting with the virus.


Assuntos
Antivirais/farmacologia , Citomegalovirus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Western Blotting , Linhagem Celular , Eletroforese em Gel de Poliacrilamida , Imunofluorescência , Humanos
6.
J Virol ; 86(6): 3370-82, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22205740

RESUMO

The product of the human cytomegalovirus (HCMV) UL71 gene is conserved throughout the herpesvirus family. During HCMV infection, protein pUL71 is required for efficient virion egress and is involved in the final steps of secondary envelopment leading to infectious viral particles. We found strong indications for oligomerization of pUL71 under native conditions when recombinant pUL71 was negatively stained and analyzed by electron microscopy. Oligomerization of pUL71 during infection was further verified by native and reducing polyacrylamide gel electrophoresis (PAGE). By in silico analyses of the pUL71 sequence, we noticed a basic leucine zipper (bZIP)-like domain, which might serve as an oligomerization domain. We demonstrated the requirement of the bZIP-like domain for pUL71 oligomerization by coimmunoprecipitation and bimolecular fluorescence complementation using a panel of pUL71 mutants. These studies revealed that the mutation of two leucine residues is sufficient to abrogate oligomerization but that intracellular localization of pUL71 was unaffected. To investigate the relevance of the bZIP domain in the viral context, recombinant viruses carrying mutations identical to those in the panel of pUL71 mutants were generated. bZIP-defective viral mutants showed impaired viral growth, a small-plaque phenotype, and an ultrastructural phenotype similar to that of the previously described UL71 stop mutant virus. The majority of virus particles within the viral assembly compartment exhibited various stages of incomplete envelopment, which is consistent with the growth defect for the bZIP mutants. From these data we conclude that the bZIP-like domain is required for oligomerization of pUL71, which seems to be essential for correct envelopment of HCMV.


Assuntos
Infecções por Citomegalovirus/virologia , Citomegalovirus/fisiologia , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/metabolismo , Montagem de Vírus , Sequência de Aminoácidos , Linhagem Celular , Citomegalovirus/química , Citomegalovirus/genética , Humanos , Zíper de Leucina , Dados de Sequência Molecular , Proteínas Estruturais Virais/genética
7.
Antiviral Res ; 206: 105402, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36007600

RESUMO

Regarding the problems with the current available drugs many research studies deal with the class of the dispirotripiperazine (DSTP)-based compounds. These are small molecules consisting of polycyclic saturated ring systems with positively charged nitrogen atoms. These compounds can interact with negatively charged HSPGs and thus block viral attachment. In a previous paper by Adfeldt et al. (2021), we have shown that the diazadispiroalkane derivatives 11826091 and 11826236 exhibit dose-dependent antiviral activity against human cytomegalovirus (HCMV) and pseudorabies virus (PrV). In the present study, these two small molecules are evaluated against two other herpesvirus species, murine cytomegalovirus (MCMV) and herpes simplex virus type 1 (HSV-1), as well as a HCMV clinical isolate. They exhibit potent antiherpetic activity against these herpesviruses with a high selectivity index. The low cytotoxicity was underlined by the LD50 determination in mice. We have shown that inhibition occurs at an early stage of infection. Interestingly, 11826091 and 11826236 reduced immediate early gene expression in HCMV and HSV-1 infected cells in a dose-dependent manner. Both small molecules probably interact electrostatically with sulfated glycosaminoglycans (GAGs) of proteoglycans on target cells resulting in blockage of adsorption sites for herpesvirus glycoprotein. Moreover, both compounds showed significant effects against the cell-associated viral spread of HSV-1 and HCMV. Overall, this study shows that 11826091 and 11826236 represent two promising candidates for a new approach of a broad antiviral therapy.


Assuntos
Herpesviridae , Herpesvirus Humano 1 , Herpesvirus Suídeo 1 , Animais , Antivirais/farmacologia , Citomegalovirus , Humanos , Camundongos
8.
Viruses ; 14(2)2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-35216006

RESUMO

Coronaviruses (CoVs) are common among humans and many animals, causing respiratory or gastrointestinal diseases. Currently, only a few antiviral drugs against CoVs are available. Especially for SARS-CoV-2, new compounds for treatment of COVID-19 are urgently needed. In this study, we characterize the antiviral effects of two high-sulfated glycosaminoglycan (GAG) derivatives against SARS-CoV-2 and bovine coronaviruses (BCoV), which are both members of the Betacoronavirus genus. The investigated compounds are based on hyaluronan (HA) and chondroitin sulfate (CS) and exhibit a strong inhibitory effect against both CoVs. Yield assays were performed using BCoV-infected PT cells in the presence and absence of the compounds. While the high-sulfated HA (sHA3) led to an inhibition of viral growth early after infection, high-sulfated CS (sCS3) had a slightly smaller effect. Time of addition assays, where sHA3 and sCS3 were added to PT cells before, during or after infection, demonstrated an inhibitory effect during all phases of infection, whereas sHA3 showed a stronger effect even after virus absorbance. Furthermore, attachment analyses with prechilled PT cells revealed that virus attachment is not blocked. In addition, sHA3 and sCS3 inactivated BCoV by stable binding. Analysis by quantitative real-time RT PCR underlines the high potency of the inhibitors against BCoV, as well as B.1-lineage, Alpha and Beta SARS-CoV-2 viruses. Taken together, these results demonstrated that the two high-sulfated GAG derivatives exhibit low cytotoxicity and represent promising candidates for an anti-CoV therapy.


Assuntos
Antivirais/farmacologia , Infecções por Coronavirus/veterinária , Coronavirus Bovino/efeitos dos fármacos , Glicosaminoglicanos/farmacologia , SARS-CoV-2/efeitos dos fármacos , Animais , Bovinos , Linhagem Celular , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacologia , Infecções por Coronavirus/tratamento farmacológico , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Humanos , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Sulfatos/química , Sulfatos/farmacologia , Ligação Viral/efeitos dos fármacos , Tratamento Farmacológico da COVID-19
9.
Viruses ; 13(3)2021 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-33809292

RESUMO

Human cytomegalovirus (HCMV), a member of the betaherpesvirinae, can cause life-threatening diseases. HCMV is globally widespread, with a seroprevalence in adults varying from 50 to 100%. HCMV infection is rarely of significant consequence in immunocompetent individuals. However, although immune control is efficient, it cannot achieve the clearance of the virus. HCMV persists lifelong in the infected host and reactivates in certain circumstances. In neonates and in immunocompromised adults, HCMV is a serious pathogen that can cause fatal organ damage. Different antiviral compounds alone or in combination have been used for the treatment of HCMV diseases. In clinical use, mutations in the viral DNA polymerase or the terminase confer resistance to ganciclovir, foscarnet, cidofovir, and letermovir. There is an urgent need to find new well-tolerated compounds supporting different modes of action. The list of novel small molecules that might have anti-HCMV activity has grown in recent years. In this short review, a selection of compounds in clinical trials and novel inhibitors targeting host-cell factors or viral proteins is presented, and their modes of action, described.


Assuntos
Antivirais/farmacologia , Infecções por Citomegalovirus/tratamento farmacológico , Citomegalovirus/efeitos dos fármacos , Proteínas Virais/antagonistas & inibidores , Descoberta de Drogas , Humanos , Hospedeiro Imunocomprometido , Replicação Viral/efeitos dos fármacos
10.
Viruses ; 13(1)2021 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-33435377

RESUMO

Human cytomegalovirus (HCMV) has been implicated in the development of human malignancies, for instance in colon cancer. Proteasome inhibitors were developed for cancer therapy and have also been shown to influence HCMV infection. The aim of this study was to investigate if proteasome inhibitors have therapeutic potential for colon carcinoma and how this is influenced by HCMV infection. We show by immunofluorescence and flow cytometry that the colon carcinoma cell line Caco-2 is susceptible to HCMV infection. Growth curve analysis as well as protein expression kinetics and quantitative genome analysis further confirm these results. HCMV has an anti-apoptotic effect on Caco-2 cells by inhibiting very early events of the apoptosis cascade. Further investigations showed that HCMV stabilizes the membrane potential of the mitochondria, which is typically lost very early during apoptosis. This stabilization is resistant to proteasome inhibitor Bortezomib treatment, allowing HCMV-infected cells to survive apoptotic signals. Our findings indicate a possible role of proteasome inhibitors in colon carcinoma therapy.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Bortezomib/farmacologia , Infecções por Citomegalovirus/metabolismo , Infecções por Citomegalovirus/virologia , Citomegalovirus/fisiologia , Células CACO-2 , Morte Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Imunofluorescência , Genoma Humano , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Potencial da Membrana Mitocondrial , Inibidores de Proteassoma/farmacologia
11.
Eur J Med Chem ; 211: 113014, 2021 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-33218683

RESUMO

Viruses are obligate intracellular parasites and have evolved to enter the host cell. To gain access they come into contact with the host cell through an initial adhesion, and some viruses from different genus may use heparan sulfate proteoglycans for it. The successful inhibition of this early event of the infection by synthetic molecules has always been an attractive target for medicinal chemists. Numerous reports have yielded insights into the function of compounds based on the dispirotripiperazine scaffold. Analysis suggests that this is a structural requirement for inhibiting the interactions between viruses and cell-surface heparan sulfate proteoglycans, thus preventing virus entry and replication. This review summarizes our current knowledge about the early history of development, synthesis, structure-activity relationships and antiviral evaluation of dispirotripiperazine-based compounds and where they are going in the future.


Assuntos
Antivirais/farmacologia , Desenho de Fármacos , Piperazinas/farmacologia , Compostos de Espiro/farmacologia , Vírus/efeitos dos fármacos , Antivirais/química , Proteoglicanas de Heparan Sulfato/antagonistas & inibidores , Proteoglicanas de Heparan Sulfato/metabolismo , Estrutura Molecular , Piperazinas/química , Compostos de Espiro/química , Vírus/metabolismo
12.
Antimicrob Agents Chemother ; 53(12): 5095-101, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19786605

RESUMO

Recently we characterized two inhibitors targeting the human cytomegalovirus (HCMV) terminase, 2-bromo-4,5,6-trichloro-1-(2,3,5-tri-O-acetyl-beta-D-ribofuranosyl) benzimidazole (BTCRB) and 2,4,5,6-tetrachloro-1-(2,3,5-tri-O-acetyl-beta-D-ribofuranosyl) benzimidazole (Cl(4)RB). The terminase consists of the ATP-hydrolyzing subunit pUL56 and the subunit pUL89 required for duplex nicking. Because mammalian cell DNA replication does not involve cleavage of concatemeric DNA by a terminase, these compounds represent attractive alternative HCMV antivirals. We now have tested these previously identified benzimidazole ribonucleosides in order to determine if they are active against HCMV clinical isolates as well as those of herpes simplex virus type 1, mouse cytomegalovirus, rat cytomegalovirus (RCMV), and varicella-zoster virus (VZV). Antiviral activity was quantified by measurement of viral plaque formation (plaque reduction) and by viral growth kinetics. Interestingly, both BTCRB and Cl(4)RB had an inhibitory effect in ganciclovir (GCV)-sensitive and GCV-resistant clinical isolates, with the best effect produced by Cl(4)RB. Electron microscopy revealed that in cells infected with GCV-sensitive or GCV-resistant isolates, B capsids and dense bodies were formed mainly. Furthermore, pulsed-field gel electrophoresis showed that cleavage of concatenated DNA was inhibited in clinical isolates. In addition, the antiviral effect on other herpesviruses was determined. Interestingly, in plaque reduction assays, BTCRB was active against all tested herpesviruses. The best effects were observed on VZV- and RCMV-infected cells. These results demonstrate that the new compounds are highly active against GCV-resistant and GCV-sensitive clinical isolates and slightly active against other herpesviruses.


Assuntos
Antivirais/farmacologia , Benzimidazóis/farmacologia , Citomegalovirus/efeitos dos fármacos , Hidrocarbonetos Halogenados/farmacologia , Ribonucleosídeos/farmacologia , Animais , Antivirais/efeitos adversos , Antivirais/química , Benzimidazóis/efeitos adversos , Benzimidazóis/química , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citomegalovirus/isolamento & purificação , Citomegalovirus/ultraestrutura , Infecções por Citomegalovirus/virologia , Eletroforese em Gel de Campo Pulsado , Herpes Simples/virologia , Herpes Zoster/virologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 3/efeitos dos fármacos , Humanos , Hidrocarbonetos Halogenados/efeitos adversos , Hidrocarbonetos Halogenados/química , Camundongos , Microscopia Eletrônica de Transmissão , Células NIH 3T3 , Ratos , Ribonucleosídeos/efeitos adversos , Ribonucleosídeos/química
13.
J Gen Virol ; 90(Pt 10): 2381-2385, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19587135

RESUMO

In order for human cytomegalovirus (HCMV) to replicate, concatemeric DNA has to be cleaved into unit-length genomes and packaged into preformed capsids. For packaging to take place and DNA to be translocated, a channel is required in the capsid. Viral capsid channels are generally formed by portal proteins. Here, we show by cross-linking, native gel electrophoresis of infected cells and gel permeation chromatography that the HCMV portal candidate protein pUL104 can form dimers and higher order multimers. Electron microscopy of purified monomeric pUL104 after 5 min incubation revealed that the protein had assembled into a multimeric form and that this form closely resembles complete portal assembly. This is the first study to show that pUL104 monomers have the ability to form portal complexes without additional viral proteins.


Assuntos
Citomegalovirus/genética , Citomegalovirus/fisiologia , Proteínas Virais/química , Proteínas Virais/fisiologia , Montagem de Vírus/fisiologia , Animais , Células Cultivadas , Cromatografia em Gel , Citomegalovirus/classificação , Eletroforese em Gel Bidimensional , Regulação Viral da Expressão Gênica/fisiologia , Humanos , Insetos/citologia , Conformação Proteica , Proteínas Virais/genética , Proteínas Virais/ultraestrutura
14.
Antiviral Res ; 161: 116-124, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30472161

RESUMO

A key step in the replication of human cytomegalovirus (HCMV) in the host cell is the generation and packaging of unit-length genomes into preformed capsids. Enzymes required for this process are so-called terminases, first described for double-stranded DNA bacteriophages. The HCMV terminase consists of the two subunits, the ATPase pUL56 and the nuclease pUL89, and a potential third component pUL51. The terminase subunits are essential for virus replication and are highly conserved throughout the Herpesviridae family. Together with the portal protein pUL104 they form a powerful biological nanomotor. It has been shown for tailed dsDNA bacteriophages that DNA translocation into preformed capsid needs an extraordinary amount of energy. The HCMV terminase subunit pUL56 provides the required ATP hydrolyzing activity. The necessary nuclease activity to cleave the concatemers into unit-length genomes is mediated by the terminase subunit pUL89. Whether this cleavage is mediated by site-specific duplex nicking has not been demonstrated, however, it is required for packaging. Binding to the portal is a prerequisite for DNA translocation. To date, it is a common view that during translocation the terminase moves along some domains of the DNA by a binding and release mechanism. These critical structures have proven to be outstanding targets for drugs to treat HCMV infections because corresponding structures do not exist in mammalian cells. Herein we examine the HCMV terminase as a target for drugs and review several inhibitors discovered by both lead-directed medicinal chemistry and by target-specific design. In addition to producing clinically active compounds the research also has furthered the understanding of the role and function of the terminase itself.


Assuntos
Antivirais/farmacologia , Infecções por Citomegalovirus/prevenção & controle , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/enzimologia , Endodesoxirribonucleases/antagonistas & inibidores , Acetatos/uso terapêutico , Animais , Ensaios Clínicos como Assunto , Citomegalovirus/genética , Infecções por Citomegalovirus/tratamento farmacológico , Humanos , Quinazolinas/uso terapêutico , Proteínas Virais/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos
15.
FEBS Lett ; 582(5): 666-72, 2008 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-18242185

RESUMO

This study provides evidence that proteasomal activity is required at multiple steps in human cytomegalovirus replication. Electron microscopy revealed that no viral particles were assembled in the presence of proteasome inhibitor MG132. Immunofluorescence and Western blot analyses using MG132 demonstrated that immediate early gene expression was suppressed at low but not high MOI. In contrast, expression of late proteins was completely blocked independent of MOI. Additionally, pulsed-field gel electrophoresis demonstrated that MG132 interferes with cleavage of HCMV DNA. Bromodeoxyuridine incorporation studies showed that de novo viral DNA synthesis is reduced in the presence of MG132. Furthermore, in contrast to previous hypotheses we demonstrated that neither the ND10 components PML and hDaxx nor NFkappaB activation represent the target for MG132.


Assuntos
Citomegalovirus/efeitos dos fármacos , Citomegalovirus/fisiologia , DNA Viral/biossíntese , Leupeptinas/farmacologia , Inibidores de Proteassoma , Montagem de Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Antivirais/farmacologia , Células Cultivadas , Infecções por Citomegalovirus/virologia , DNA Concatenado/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/virologia , Humanos , NF-kappa B/deficiência , NF-kappa B/metabolismo , Proteínas Virais/metabolismo , Vírion/efeitos dos fármacos , Vírion/metabolismo
16.
Antiviral Res ; 137: 102-107, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27871886

RESUMO

BACKGROUND: Benzimidazole D-ribonucleosides are potent and selective inhibitors of CMV infection that have been shown to target the viral terminase, the enzyme complex responsible for viral DNA cleavage into single unit-length genomes and subsequent DNA packaging into procapsids. Here, we evaluated the viral inhibition by benzimidazole D-ribonucleosides against rat cytomegalovirus (RCMV). METHODS: Antiviral activity of compounds Cl4RB and BTCRB against RCMV was quantified by measurement of plaque formation. Yield assays and electron microscopy of thin sections was performed using RCMV-infected cells in the presence or absence of the compounds. The effects of Cl4RB and BTCRB on cleavage of concatemers was analyzed by pulsed-field gel electrophoresis. To characterize the behaviour of the antiviral compounds in a more physiological environment, a 3D cell culture model was employed where cells are embedded in an extracellular matrix using rat-tail collagen I. RESULTS: Both compounds had an inhibitory effect against RCMV-E. Electron microscopy revealed that only few virions were formed in RCMV-E infected cells in the presence of the compounds. Pulsed-field gel electrophoresis showed that DNA concatemers failed to be processed in the presence of the compounds. Yield Assays showed a comparable viral growth in the 3D vs. 2D cell culture as well as inhibition in the presence of Cl4RB or BTCRB for RCMV-E/GFP. CONCLUSIONS: These results demonstrate that the tetrahalogenated benzimidazole D-ribonucleosides are effective against RCMV-E by preventing cleavage of concatemeric DNA and nuclear egress of mature capsids.


Assuntos
Antivirais/farmacologia , Benzimidazóis/farmacologia , Infecções por Herpesviridae/tratamento farmacológico , Muromegalovirus/efeitos dos fármacos , Nucleosídeos/farmacologia , Ribonucleosídeos/farmacologia , Animais , Antivirais/química , Benzimidazóis/química , Técnicas de Cultura de Células , Colágeno/química , Empacotamento do DNA/efeitos dos fármacos , Endodesoxirribonucleases/efeitos dos fármacos , Halogenação , Infecções por Herpesviridae/virologia , Microscopia Eletrônica , Modelos Biológicos , Muromegalovirus/ultraestrutura , Nucleosídeos/química , Ratos , Ribonucleosídeos/química , Alicerces Teciduais , Ensaio de Placa Viral
17.
Biochemistry ; 45(51): 15259-68, 2006 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-17176048

RESUMO

Terminase enzymes are common to complex double-stranded DNA viruses and function to package viral DNA into the capsid. We recently demonstrated that the bacteriophage lambda terminase gpA and gpNu1 proteins assemble into a stable heterotrimer with a molar ratio gpA1/gpNu1(2). This terminase protomer possesses DNA maturation and packaging activities that are dependent on the E. coli integration host factor protein (IHF). Here, we show that the protomer further assembles into a homogeneous tetramer of protomers of composition (gpA1/gpNu1(2))4. Electron microscopy shows that the tetramer forms a ring structure large enough to encircle duplex DNA. In contrast to the heterotrimer, the ring tetramer can mature and package viral DNA in the absence of IHF. We propose that IHF induced bending of viral DNA facilitates the assembly of four terminase protomers into a ring tetramer that represents the catalytically competent DNA maturation and packaging complex in vivo. This work provides, for the first time, insight into the functional assembly state of a viral DNA packaging motor.


Assuntos
Bacteriófago lambda/enzimologia , Replicação do DNA/genética , DNA Viral/biossíntese , Endodesoxirribonucleases/química , Endodesoxirribonucleases/metabolismo , Proteínas Motores Moleculares/química , Proteínas Motores Moleculares/metabolismo , Montagem de Vírus/fisiologia , Bacteriófago lambda/genética , Bacteriófago lambda/fisiologia , Bacteriófago lambda/ultraestrutura , Catálise , DNA Viral/química , DNA Viral/ultraestrutura , Endodesoxirribonucleases/ultraestrutura , Proteínas Motores Moleculares/ultraestrutura , Peso Molecular , Regiões Promotoras Genéticas , Ultracentrifugação
18.
FEBS Lett ; 580(26): 6132-8, 2006 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-17056039

RESUMO

To clearly demonstrate the role of the human cytomegalovirus (HCMV) portal protein pUL104 RNA interference was applied. Expressing cell lines were constructed by transduction of shRNAs via the infection with retroviral vectors. After infection of these cells with HCMV AD169 the expression of pUL104 was reduced up to 80% for shRNA S1 and 54% for shRNA S2 at late times of infection compared to controls. In addition, the inhibitory effect was corresponding with a decrease in viral mRNA and plaque formations. Electron microscopic analysis demonstrated that infection of cells expressing pUL104-specific shRNAs resulted in the inhibition of formation of replicative particles.


Assuntos
Citomegalovirus/fisiologia , RNA Interferente Pequeno/farmacologia , RNA Viral/efeitos dos fármacos , Montagem de Vírus/efeitos dos fármacos , Linhagem Celular , Citomegalovirus/efeitos dos fármacos , Infecções por Citomegalovirus/tratamento farmacológico , Humanos , Interferência de RNA , Ensaio de Placa Viral , Vírion/efeitos dos fármacos
19.
Nucleic Acids Res ; 31(5): 1426-33, 2003 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-12595550

RESUMO

Human cytomegalovirus (HCMV) terminase is composed of subunits pUL56 (130 kDa) and pUL89 ( approximately 75 kDa), encoded by the UL56 and UL89 genes. In a recent investigation, we demonstrated that the main ATPase activity is associated with the large terminase subunit pUL56. The protein has two putative ATP-binding sites, which were suggested to be composed of the sequence (amino acids 463-470) for ATP-binding site 1 and YNETFGKQ (amino acids 709-716) for the second site. We now demonstrate using a 1.5 kb fragment encoding the C-terminal half of pUL56 that ATP-binding site 1 is not critical for the function, whereas ATP-binding site 2 is required for the enzymatic activity. Mutation G714A in this protein reduced the ATPase activity to approximately 65% and the double mutation G714A/K715N showed a reduction up to 75%. However, the substitution of E711A revoked the effect of the substitutions. The functional character of the ATP-binding site was demonstrated by transfer of YNETFGKQLSIACLR (709-723) to glutathione-S-transferase (GST). Interestingly, vanadate, an ATPase inhibitor, has the ability to block the ATPase activity of pUL56 as well as of Apyrase, while the antitumor ATP-mimetic agent geldanamycin, did not affect the ATP-binding of pUL56. Furthermore, in contrast to an inactive control compound, the specific HCMV terminase inhibitor BDCRB showed a partial inhibition of the pUL56-specific ATPase activity. Our results clearly demonstrated that (i) the enzymatic activity of the terminase subunit pUL56 could be inhibited by vanadate, (ii) only the ATP-binding site 2 is critical for the pUL56 function and (iii) glycine G714 is an invariant amino acid.


Assuntos
Trifosfato de Adenosina/metabolismo , Citomegalovirus/enzimologia , Endodesoxirribonucleases/metabolismo , Adenosina Trifosfatases/antagonistas & inibidores , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Benzoquinonas , Sítios de Ligação/genética , Endodesoxirribonucleases/genética , Inibidores Enzimáticos/farmacologia , Lactamas Macrocíclicas , Dados de Sequência Molecular , Mutação , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Quinonas/farmacologia , Homologia de Sequência de Aminoácidos , Vanadatos/farmacologia
20.
Nucleic Acids Res ; 30(7): 1695-703, 2002 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11917032

RESUMO

Herpesvirus DNA packaging involves binding and cleavage of DNA containing the specific DNA-packaging motifs. Here we report a first characterization of the terminase subunits pUL56 and pUL89 of human cytomegalovirus (HCMV). Both gene products were shown to have comparable nuclease activities in vitro. Under limiting protein concentrations the nuclease activity is enhanced by interaction of pUL56 and pUL89. High amounts of 2-bromo-5,6-dichloro-1-beta-D-ribofuranosyl benzimidazole partially inhibited the pUL89-associated nuclease activity. It was demonstrated that pUL56 is able to bind to nucleocapsids in vivo. Electron microscopy (EM) and image analysis of purified pUL56 revealed that the molecules occurred as a distinct ring-shaped structure with a pronounced cleft. EM analysis of purified pUL89 demonstrated that this protein is also a toroidal DNA-metabolizing protein. Upon interaction of pUL56 with linearized DNA, the DNA remains uncut while the cutting event itself is mediated by pUL89. Using biochemical assays in conjunction with EM pUL56 was shown to (i) bind to DNA and (ii) associate with the capsid. In contrast to this, EM analysis implied that pUL89 is required to effect DNA cleavage. The data provide the first insights into the terminase-dependent viral DNA-packaging mechanism of HCMV.


Assuntos
Citomegalovirus/enzimologia , DNA/metabolismo , Endodesoxirribonucleases/metabolismo , Animais , Antivirais/farmacologia , Benzimidazóis/farmacologia , Capsídeo/metabolismo , Linhagem Celular , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/genética , DNA/genética , Desoxirribonucleases/efeitos dos fármacos , Desoxirribonucleases/metabolismo , Relação Dose-Resposta a Droga , Endodesoxirribonucleases/genética , Endodesoxirribonucleases/ultraestrutura , Microscopia Eletrônica , Subunidades Proteicas , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Ribonucleosídeos/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA