Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Viruses ; 13(3)2021 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-33799898

RESUMO

Herpesviral nuclear egress is a regulated process shared by all family members, ensuring the efficient cytoplasmic release of viral capsids. In the case of human cytomegalovirus (HCMV), the core of the nuclear egress complex (NEC) consists of the pUL50-pUL53 heterodimer that builds hexameric lattices for capsid binding and multicomponent interaction, including NEC-associated host factors. A characteristic feature of NEC interaction is the N-terminal hook structure of pUL53 that binds to an alpha-helical groove of pUL50, thus termed as hook-into-groove interaction. This central regulatory element is essential for viral replication and shows structural-functional conservation, which has been postulated as a next-generation target of antiviral strategies. However, a solid validation of this concept has been missing. In the present study, we focused on the properties of oligomeric HCMV core NEC interaction and the antiviral activity of specifically targeted prototype inhibitors. Our data suggest the following: (i) transiently expressed, variably tagged versions of HCMV NEC proteins exert hook-into-groove complexes, putatively in oligomeric assemblies that are distinguishable from heterodimers, as shown by in vitro assembly and coimmunoprecipitation approaches; (ii) this postulated oligomeric binding pattern was further supported by the use of a pUL50::pUL53 fusion construct also showing a pronounced multi-interaction potency; (iii) using confocal imaging cellular NEC-associated proteins were found partly colocalized with the tagged core NECs; (iv) a small inhibitory molecule, recently identified by an in vitro binding inhibition assay, was likewise active in blocking pUL50-pUL53 oligomeric assembly and in exerting antiviral activity in HCMV-infected fibroblasts. In summary, the findings refine the previous concept of HCMV core NEC formation and nominate this drug-accessible complex as a validated antiviral drug target.


Assuntos
Antivirais/farmacologia , Infecções por Citomegalovirus/tratamento farmacológico , Citomegalovirus/crescimento & desenvolvimento , Proteínas Virais/metabolismo , Liberação de Vírus/efeitos dos fármacos , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Núcleo Celular/virologia , Citomegalovirus/efeitos dos fármacos , Infecções por Citomegalovirus/patologia , Células HEK293 , Células HeLa , Humanos , Proteínas de Membrana/metabolismo , Simulação de Dinâmica Molecular , Membrana Nuclear/virologia , Ligação Proteica
2.
Viruses ; 13(3)2021 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-33809234

RESUMO

Nuclear egress is an essential process in the replication of human cytomegalovirus (HCMV), as it enables the migration of newly formed viral capsids from the nucleus into the cytoplasm. Inhibition of the HCMV core nuclear egress complex (core NEC), composed of viral proteins pUL50 and pUL53, has been proposed as a potential new target for the treatment of HCMV infection and disease. Here, we present a new type of small molecule inhibitors of HCMV core NEC formation, which inhibit the pUL50-pUL53 interaction at nanomolar concentrations. These inhibitors, i.e., verteporfin and merbromin, were identified through the screening of the Prestwick Chemical Library® of approved drug compounds. The inhibitory effect of merbromin is both compound- and target-specific, as no inhibition was seen for other mercury-organic compounds. Furthermore, merbromin does not inhibit an unrelated protein-protein interaction either. More importantly, merbromin was found to inhibit HCMV infection of cells in three different assays, as well as to disrupt HCMV NEC nuclear rim formation. Thus, while not being an ideal drug candidate by itself, merbromin may serve as a blueprint for small molecules with high HCMV core NEC inhibitory potential, as candidates for novel anti-herpesviral drugs.


Assuntos
Antivirais/farmacologia , Infecções por Citomegalovirus/virologia , Citomegalovirus/metabolismo , Merbromina/farmacologia , Proteínas Virais/metabolismo , Vírion/metabolismo , Células Cultivadas , Fibroblastos , Humanos , Cultura Primária de Células , Liberação de Vírus , Replicação Viral
3.
PLoS Pathog ; 17(3): e1009471, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33780515

RESUMO

Platelet-derived growth factor receptor alpha (PDGFRα) serves as an entry receptor for the human cytomegalovirus (HCMV), and soluble PDGFRα-Fc can neutralize HCMV at a half-maximal effective concentration (EC50) of about 10 ng/ml. While this indicates a potential for usage as an HCMV entry inhibitor PDGFRα-Fc can also bind the physiological ligands of PDGFRα (PDGFs), which likely interferes with the respective signaling pathways and represents a potential source of side effects. Therefore, we tested the hypothesis that interference with PDGF signaling can be prevented by mutations in PDGFRα-Fc or combinations thereof, without losing the inhibitory potential for HCMV. To this aim, a targeted mutagenesis approach was chosen. The mutations were quantitatively tested in biological assays for interference with PDGF-dependent signaling as well as inhibition of HCMV infection and biochemically for reduced affinity to PDGF-BB, facilitating quantification of PDGFRα-Fc selectivity for HCMV inhibition. Mutation of Ile 139 to Glu and Tyr 206 to Ser strongly reduced the affinity for PDGF-BB and hence interference with PDGF-dependent signaling. Inhibition of HCMV infection was less affected, thus increasing the selectivity by factor 4 and 8, respectively. Surprisingly, the combination of these mutations had an additive effect on binding of PDGF-BB but not on inhibition of HCMV, resulting in a synergistic 260fold increase of selectivity. In addition, a recently reported mutation, Val 242 to Lys, was included in the analysis. PDGFRα-Fc with this mutation was fully effective at blocking HCMV entry and had a drastically reduced affinity for PDGF-BB. Combining Val 242 to Lys with Ile 139 to Glu and/or Tyr 206 to Ser further reduced PDGF ligand binding beyond detection. In conclusion, this targeted mutagenesis approach identified combinations of mutations in PDGFRα-Fc that prevent interference with PDGF-BB but maintain inhibition of HCMV, which qualifies such mutants as candidates for the development of HCMV entry inhibitors.


Assuntos
Infecções por Citomegalovirus , Fragmentos Fc das Imunoglobulinas , Receptor alfa de Fator de Crescimento Derivado de Plaquetas , Becaplermina/efeitos dos fármacos , Becaplermina/metabolismo , Citomegalovirus , Fibroblastos , Células HEK293 , Humanos , Fragmentos Fc das Imunoglobulinas/química , Fragmentos Fc das Imunoglobulinas/farmacologia , Mutagênese Sítio-Dirigida , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/química , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/farmacologia
4.
Clin Infect Dis ; 68(8): 1420-1426, 2019 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-30137245

RESUMO

Despite advances in preventive strategies, cytomegalovirus (CMV) infection remains a major complication in solid organ and hematopoietic cell transplant recipients. CMV infection may fail to respond to commercially available antiviral therapies, with or without demonstrating genotypic mutation(s) known to be associated with resistance to these therapies. This lack of response has been termed "resistant/refractory CMV" and is a key focus of clinical trials of some investigational antiviral agents. To provide consistent criteria for future clinical trials and outcomes research, the CMV Resistance Working Group of the CMV Drug Development Forum (consisting of scientists, clinicians, regulatory officials, and industry representatives from the United States, Canada, and Europe) has undertaken establishing standardized consensus definitions of "resistant" and "refractory" CMV. These definitions have emerged from the Working Group's review of the available virologic and clinical literature and will be subject to reassessment and modification based on results of future studies.


Assuntos
Infecções por Citomegalovirus/classificação , Transplante de Células-Tronco Hematopoéticas , Transplante de Órgãos , Transplantados , Farmacorresistência Viral , Humanos , Hospedeiro Imunocomprometido , Fatores de Risco , Terminologia como Assunto , Falha de Tratamento
5.
J Virol ; 91(12)2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28356534

RESUMO

Human cytomegalovirus (HCMV) genome encapsidation requires several essential viral proteins, among them pUL56, pUL89, and the recently described pUL51, which constitute the viral terminase. To gain insight into terminase complex assembly, we investigated interactions between the individual subunits. For analysis in the viral context, HCMV bacterial artificial chromosomes carrying deletions in the open reading frames encoding the terminase proteins were used. These experiments were complemented by transient-transfection assays with plasmids expressing the terminase components. We found that if one terminase protein was missing, the levels of the other terminase proteins were markedly diminished, which could be overcome by proteasome inhibition or providing the missing subunit in trans These data imply that sequestration of the individual subunits within the terminase complex protects them from proteasomal turnover. The finding that efficient interactions among the terminase proteins occurred only when all three were present together is reminiscent of a folding-upon-binding principle leading to cooperative stability. Furthermore, whereas pUL56 was translocated into the nucleus on its own, correct nuclear localization of pUL51 and pUL89 again required all three terminase constituents. Altogether, these features point to a model of the HCMV terminase as a multiprotein complex in which the three players regulate each other concerning stability, subcellular localization, and assembly into the functional tripartite holoenzyme.IMPORTANCE HCMV is a major risk factor in immunocompromised individuals, and congenital CMV infection is the leading viral cause for long-term sequelae, including deafness and mental retardation. The current treatment of CMV disease is based on drugs sharing the same mechanism, namely, inhibiting viral DNA replication, and often results in adverse side effects and the appearance of resistant virus strains. Recently, the HCMV terminase has emerged as an auspicious target for novel antiviral drugs. A new drug candidate inhibiting the HCMV terminase, Letermovir, displayed excellent potency in clinical trials; however, its precise mode of action is not understood yet. Here, we describe the mutual dependence of the HCMV terminase constituents for their assembly into a functional terminase complex. Besides providing new basic insights into terminase formation, these results will be valuable when studying the mechanism of action for drugs targeting the HCMV terminase and developing additional substances interfering with viral genome encapsidation.


Assuntos
Citomegalovirus/enzimologia , Endodesoxirribonucleases/metabolismo , Proteínas Virais/metabolismo , Proteínas Estruturais Virais/metabolismo , Transporte Ativo do Núcleo Celular/genética , Linhagem Celular , Cromossomos Artificiais Bacterianos/genética , Citomegalovirus/genética , Citomegalovirus/metabolismo , DNA Viral , Endodesoxirribonucleases/química , Endodesoxirribonucleases/genética , Fibroblastos/virologia , Genoma Viral , Células HeLa , Humanos , Complexo de Endopeptidases do Proteassoma/genética , Estabilidade Proteica , Proteínas Virais/genética , Proteínas Estruturais Virais/genética
6.
Antiviral Res ; 132: 204-9, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27345658

RESUMO

Letermovir is a novel anti-HCMV drug in Phase III development that targets the UL56 subunit of the viral terminase complex. In immunocompromised patients four major glycoprotein B (gB) subtypes are known and may influence pathogenesis and thus disease outcomes. Using a panel of 74 letermovir-naïve, low-passage, clinical HCMV isolates, we examined the potential impact of i) gB genotype and ii) naturally occurring UL56 sequence variations upon susceptibility to letermovir. Our data show that letermovir's potency is independent of gB subtype and show that naturally-occurring letermovir-resistance is rare or possibly absent.


Assuntos
Acetatos/farmacologia , Antivirais/farmacologia , Infecções por Citomegalovirus/virologia , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/genética , Genótipo , Polimorfismo Genético , Quinazolinas/farmacologia , Proteínas do Envelope Viral/genética , Proteínas Estruturais Virais/genética , Citomegalovirus/isolamento & purificação , Farmacorresistência Viral , Ganciclovir/farmacologia , Humanos , Testes de Sensibilidade Microbiana , Mutação , Polimorfismo de Nucleotídeo Único
7.
J Infect Dis ; 213(1): 23-30, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26113373

RESUMO

BACKGROUND: The efficacy of different letermovir (AIC246, MK8228) doses (60, 120, and 240 mg/day) against human cytomegalovirus (HCMV) was evaluated in a recent phase 2b dose-range-finding prophylaxis study in stem-cell transplant recipients. Here we report the genotypic and phenotypic characterization of 15 viral breakthroughs considered to be virological failures. METHODS: Direct sequencing of an HCMV open reading frame UL56 region that included amino acids 230-370 and thus encompassed all known letermovir resistance mutations was followed by marker-transfer experiments to assess the impact of the identified sequence polymorphisms on viral fitness and susceptibility to letermovir. RESULTS: UL56 genotyping was successful for 12 of 15 patients. Six amino acid substitutions were detected in 5 patients. In 1 subject from the 60-mg-dose group, the known letermovir resistance mutation V236M was identified subsequent to a wild-type viremic episode. The remaining 5 sequence variants (L134V, S227I, Q228H, R410G, and D414N) were shown to be inert with regard to letermovir susceptibility, thus representing natural polymorphisms. CONCLUSIONS: Our findings represent the first case of a letermovir resistance mutation emerging in the clinic, apparently because of a suboptimal prophylactic dose (60 mg/day). This is in agreement with the trial's efficacy analyses, findings of which suggest that letermovir doses of 60 mg/day and 120 mg/day are suboptimal for prophylaxis whereas a dose of 240 mg/day appears to achieve complete suppression of viremia.


Assuntos
Infecções por Citomegalovirus/tratamento farmacológico , Infecções por Citomegalovirus/virologia , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/genética , Acetatos/farmacologia , Acetatos/uso terapêutico , Antivirais/farmacologia , Antivirais/uso terapêutico , Infecções por Citomegalovirus/epidemiologia , DNA Viral/sangue , DNA Viral/genética , Farmacorresistência Viral/efeitos dos fármacos , Farmacorresistência Viral/genética , Humanos , Fases de Leitura Aberta/genética , Quinazolinas/farmacologia , Quinazolinas/uso terapêutico , Carga Viral , Viremia
8.
Antimicrob Agents Chemother ; 59(6): 3140-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25779572

RESUMO

Despite modern prevention and treatment strategies, human cytomegalovirus (HCMV) remains a common opportunistic pathogen associated with serious morbidity and mortality in immunocompromised individuals, such as transplant recipients and AIDS patients. All drugs currently licensed for the treatment of HCMV infection target the viral DNA polymerase and are associated with severe toxicity issues and the emergence of drug resistance. Letermovir (AIC246, MK-8228) is a new anti-HCMV agent in clinical development that acts via a novel mode of action and has demonstrated anti-HCMV activity in vitro and in vivo. For the future, drug combination therapies, including letermovir, might be indicated under special medical conditions, such as the emergence of multidrug-resistant virus strains in transplant recipients or in HCMV-HIV-coinfected patients. Accordingly, knowledge of the compatibility of letermovir with other HCMV or HIV antivirals is of medical importance. Here, we evaluated the inhibition of HCMV replication by letermovir in combination with all currently approved HCMV antivirals using cell culture checkerboard assays. In addition, the effects of letermovir on the antiviral activities of selected HIV drugs, and vice versa, were analyzed. Using two different mathematical techniques to analyze the experimental data, (i) additive effects were observed for the combination of letermovir with anti-HCMV drugs and (ii) no interaction was found between letermovir and anti-HIV drugs. Since none of the tested drug combinations significantly antagonized letermovir efficacy (or vice versa), our findings suggest that letermovir may offer the potential for combination therapy with the tested HCMV and HIV drugs.


Assuntos
Acetatos/farmacologia , Fármacos Anti-HIV/farmacologia , Antivirais/farmacologia , Citomegalovirus/efeitos dos fármacos , Quinazolinas/farmacologia , Linhagem Celular , Combinação de Medicamentos , Farmacorresistência Viral , Humanos , Replicação Viral/efeitos dos fármacos
9.
Antiviral Res ; 116: 48-50, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25637709

RESUMO

Letermovir is a new drug in Phase 3 clinical development for the prevention of human Cytomegalovirus (HCMV) infections in hematopoietic-stem-cell transplant recipients (HSCT). In contrast to marketed anti-HCMV drugs which all target the viral DNA polymerase, letermovir's novel mode of action targets the UL56 subunit of the viral terminase complex. Consistently letermovir resistance has mapped in vitro to a distinct region within ORF UL56 (amino acid 230-370). Here we used marker transfer to demonstrate that two naturally occurring UL56 sequence variants within this region, located directly adjacent to sites known to mediate letermovir resistance in vitro (D242G and A327V) represent normal interstrain polymorphisms unrelated to drug-resistance.


Assuntos
Acetatos/farmacologia , Antivirais/farmacologia , Infecções por Citomegalovirus/tratamento farmacológico , Citomegalovirus/genética , Farmacorresistência Viral/genética , Polimorfismo Genético , Quinazolinas/farmacologia , Proteínas Estruturais Virais/genética , Citomegalovirus/efeitos dos fármacos , Infecções por Citomegalovirus/virologia , Genótipo , Transplante de Células-Tronco Hematopoéticas , Humanos , Mutação , Fases de Leitura Aberta , Fenótipo
10.
N Engl J Med ; 370(19): 1781-9, 2014 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-24806159

RESUMO

BACKGROUND: Cytomegalovirus (CMV) infection is a leading cause of illness and death in patients who have undergone allogeneic hematopoietic-cell transplantation. Available treatments are restricted by clinically significant toxic effects and drug resistance. METHODS: In this phase 2 study, we evaluated the effect of letermovir (also known as AIC246), a new anti-CMV drug with a novel mechanism of action, on the incidence and time to onset of prophylaxis failure in CMV-seropositive recipients of allogeneic hematopoietic-cell transplants from matched related or unrelated donors. From March 2010 through October 2011, we randomly assigned 131 transplant recipients in a 3:1 ratio to three sequential study cohorts according to a double-blind design. Patients received oral letermovir (at a dose of 60, 120, or 240 mg per day, or matching placebo) for 12 weeks after engraftment. The primary end point was all-cause prophylaxis failure, defined as discontinuation of the study drug because of CMV antigen or DNA detection, end-organ disease, or any other cause. Patients underwent weekly surveillance for CMV infection. RESULTS: The reduction in the incidence of all-cause prophylaxis failure was dose-dependent. The incidence of prophylaxis failure with letermovir, as compared with placebo, was 48% versus 64% at a daily letermovir dose of 60 mg (P=0.32), 32% at a dose of 120 mg (P=0.01), and 29% at a dose of 240 mg (P=0.007). Kaplan-Meier time-to-onset profiles for prophylaxis failure showed a significant difference in the comparison of letermovir at a dose of 240 mg per day with placebo (P=0.002). The safety profile of letermovir was similar to placebo, with no indication of hematologic toxicity or nephrotoxicity. CONCLUSIONS: Letermovir, as compared with placebo, was effective in reducing the incidence of CMV infection in recipients of allogeneic hematopoietic-cell transplants. The highest dose (240 mg per day) had the greatest anti-CMV activity, with an acceptable safety profile. (Funded by AiCuris; ClinicalTrials.gov number, NCT01063829.).


Assuntos
Acetatos/administração & dosagem , Antivirais/administração & dosagem , Infecções por Citomegalovirus/prevenção & controle , Transplante de Células-Tronco Hematopoéticas , Hospedeiro Imunocomprometido , Infecções Oportunistas/prevenção & controle , Quinazolinas/administração & dosagem , Acetatos/efeitos adversos , Adulto , Antivirais/efeitos adversos , Relação Dose-Resposta a Droga , Método Duplo-Cego , Humanos , Incidência , Estimativa de Kaplan-Meier , Quinazolinas/efeitos adversos , Transplante Homólogo , Falha de Tratamento
11.
Antimicrob Agents Chemother ; 58(1): 610-3, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24189264

RESUMO

Letermovir is a novel antiviral compound currently in clinical development for the prevention of human cytomegalovirus (HCMV) infections. In contrast to all currently approved anti-HCMV drugs that target the viral DNA polymerase, letermovir acts via a distinct mode of action involving the viral terminase subunit pUL56. To extend our understanding of potential letermovir resistance mechanisms, we used marker transfer to characterize mutations identified in letermovir-resistant HCMV variants that were selected in cell culture.


Assuntos
Acetatos/farmacologia , Antivirais/farmacologia , Citomegalovirus/genética , Quinazolinas/farmacologia , Citomegalovirus/efeitos dos fármacos , DNA Polimerase Dirigida por DNA/genética , Farmacorresistência Viral/genética , Humanos
12.
Transpl Int ; 27(1): 77-86, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24164420

RESUMO

Cytomegalovirus (CMV) infection remains a significant cause of morbidity and mortality in transplant recipients. Letermovir (AIC246), is a novel anti-HCMV drug in development, acting via a novel mechanism of action. In this proof-of-concept trial with first administration of letermovir to patients, 27 transplant recipients with active CMV replication were randomly assigned to a 14-day oral treatment regimen of either letermovir 40 mg twice a day, letermovir 80 mg once a day, or local standard of care (SOC) in a multicenter, open-label trial. Efficacy, safety, and limited pharmacokinetic parameters were assessed. All groups had a statistically significant decrease in CMV-DNA copy number from baseline (40 mg BID: P = 0.031; 80 mg QD: P = 0.018; SOC: P = 0.001), and comparison of viral load reduction between treatment groups showed no statistically significant differences. Viral clearance was achieved for 6 of 12 patients (50%) in the letermovir groups versus two of seven SOC patients (28.6%). Letermovir treatment was generally well tolerated, no patient developed CMV disease during the trial. Both letermovir treatment regimens resulted in equally high trough level plasma concentrations. The efficacy, safety, and pharmacokinetics observed in these viremic transplant recipients indicate that letermovir is a promising new anti-CMV drug.


Assuntos
Acetatos/administração & dosagem , Infecções por Citomegalovirus/tratamento farmacológico , Transplante de Rim , Quinazolinas/administração & dosagem , Acetatos/farmacocinética , Adulto , Idoso , Antivirais/administração & dosagem , Antivirais/farmacocinética , Citomegalovirus/genética , Infecções por Citomegalovirus/virologia , DNA Viral/sangue , Feminino , Humanos , Transplante de Rim/efeitos adversos , Masculino , Pessoa de Meia-Idade , Infecções Oportunistas/tratamento farmacológico , Infecções Oportunistas/virologia , Quinazolinas/farmacocinética , Carga Viral , Viremia/virologia
13.
Med Microbiol Immunol ; 201(4): 567-79, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22965172

RESUMO

The phosphoprotein 65 (pp65) of human cytomegalovirus is a prominent target of the antiviral CD8 T lymphocyte response. This study focused on investigating the properties of pp65 that render it a privileged antigen. It was found that pp65 was metabolically stable. The tegument protein was introduced into MHC class I presentation following its delivery via non-replicating dense bodies. No ubiquitination was found on particle-associated pp65. Proof was obtained that pp65 was a nucleocytoplasmic shuttle protein, using heterokaryon analyses. Based on this finding, inhibition experiments showed that presentation of particle-derived pp65 by HLA-A2 was sensitive to the impairment of the CRM1-mediated nuclear export pathway. The data support the idea that particle-derived pp65 can serve as a nuclear reservoir for proteasomal processing and MHC class I presentation, following its CRM1-dependent nuclear export. The presentation of pp65-derived peptides was also impaired by CRM1-inhibition following de novo synthesis of the tegument protein. However, pp65 protein levels were also reduced when blocking CRM1-mediated export after transient expression. This indicated that pp65 expression rather than direct interference with its own nuclear export was responsible for its reduced presentation in this case. The functionality of CRM1-mediated nuclear export is thus important for the presentation of pp65-derived peptides in the context of MHC class I on organ cells, both after exogenous uptake and after de novo synthesis of the tegument protein, but different mechanisms may account for either case.


Assuntos
Apresentação de Antígeno , Antígenos Virais/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Carioferinas/metabolismo , Fosfoproteínas/imunologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas da Matriz Viral/imunologia , Transporte Ativo do Núcleo Celular , Núcleo Celular/química , Células Cultivadas , Citoplasma/química , Humanos , Proteína Exportina 1
14.
Antimicrob Agents Chemother ; 56(2): 1135-7, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22106211

RESUMO

AIC246 (letermovir) is a potent anticytomegalovirus drug in clinical development. Here, we report a consistent antiviral efficacy of AIC246 against human cytomegalovirus laboratory strains, clinical isolates, and virus variants resistant to approved drugs. Furthermore, we describe a remarkable selectivity of AIC246 for human cytomegaloviruses compared to that of other alpha-, beta-, or gammaherpesviruses or nonrelated pathogenic viruses, including adeno-, hepadna-, retro-, orthomyxo-, and flaviviruses. Our data confirm and support an excellent and selective anticytomegaloviral activity of AIC246.


Assuntos
Antivirais/farmacologia , Infecções por Citomegalovirus/virologia , Citomegalovirus/efeitos dos fármacos , Quinazolinas/farmacologia , Antivirais/química , Citomegalovirus/isolamento & purificação , Farmacorresistência Viral/efeitos dos fármacos , Herpesviridae/classificação , Herpesviridae/efeitos dos fármacos , Humanos , Testes de Sensibilidade Microbiana , Quinazolinas/química , Especificidade da Espécie , Viroses/virologia , Vírus/classificação , Vírus/efeitos dos fármacos
15.
PLoS One ; 6(7): e22671, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21799930

RESUMO

Cellular DExD/H-box RNA-helicases perform essential functions during mRNA biogenesis. The closely related human proteins UAP56 and URH49 are members of this protein family and play an essential role for cellular mRNA export by recruiting the adaptor protein REF to spliced and unspliced mRNAs. In order to gain insight into their mode of action, we aimed to characterize these RNA-helicases in more detail. Here, we demonstrate that UAP56 and URH49 exhibit an intrinsic CRM1-independent nucleocytoplasmic shuttling activity. Extensive mapping studies identified distinct regions within UAP56 or URH49 required for (i) intranuclear localization (UAP56 aa81-381) and (ii) interaction with REF (UAP56 aa51-428). Moreover, the region conferring nucleocytoplasmic shuttling activity was mapped to the C-terminus of UAP56, comprising the amino acids 195-428. Interestingly, this region coincides with a domain within Uap56p of S. pombe that has been reported to be required for both Rae1p-interaction and nucleocytoplasmic shuttling. However, in contrast to this finding we report that human UAP56 shuttles independently from Rae1. In summary, our results reveal nucleocytoplasmic shuttling as a conserved feature of yeast and human UAP56, while their export receptor seems to have diverged during evolution.


Assuntos
Núcleo Celular/metabolismo , Citoplasma/metabolismo , RNA Helicases DEAD-box/metabolismo , Carioferinas/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Transporte Ativo do Núcleo Celular , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , RNA Helicases DEAD-box/química , RNA Helicases DEAD-box/genética , Células HEK293 , Humanos , Camundongos , Dados de Sequência Molecular , Mutagênese , Mutação , Células NIH 3T3 , Proteínas Associadas à Matriz Nuclear/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Estrutura Terciária de Proteína , Proteína Exportina 1
16.
J Virol ; 85(20): 10884-93, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21752907

RESUMO

Human cytomegalovirus (HCMV) remains the leading viral cause of birth defects and life-threatening disease in transplant recipients. All approved antiviral drugs target the viral DNA polymerase and are associated with severe toxicity issues and the emergence of drug resistance. Attempts to discover improved anti-HCMV drugs led to the identification of the small-molecular-weight compound AIC246 (Letermovir). AIC246 exhibits outstanding anti-HCMV activity in vitro and in vivo and currently is undergoing a clinical phase IIb trial. The initial mode-of-action studies suggested that the drug acts late in the HCMV replication cycle via a mechanism distinct from that of polymerase inhibitors. Here, we extend our mode-of-action analyses and report that AIC246 blocks viral replication without inhibiting the synthesis of progeny HCMV DNA or viral proteins. The genotyping of mutant viruses that escaped AIC246 inhibition uncovered distinct point mutations in the UL56 subunit of the viral terminase complex. Marker transfer analyses confirmed that these mutations were sufficient to mediate AIC246 resistance. The mapping of drug resistance to open reading frame UL56 suggests that viral DNA processing and/or packaging is targeted by AIC246. In line with this, we demonstrate that AIC246 affects the formation of proper unit-length genomes from viral DNA concatemers and interferes with virion maturation. However, since AIC246-resistant viruses do not exhibit cross-resistance to previously published terminase inhibitors, our data suggest that AIC246 interferes with HCMV DNA cleavage/packaging via a molecular mechanism that is distinct from that of other compound classes known to target the viral terminase.


Assuntos
Antivirais/farmacologia , Citomegalovirus/efeitos dos fármacos , Endodesoxirribonucleases/metabolismo , Proteínas Estruturais Virais/metabolismo , Montagem de Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Análise Mutacional de DNA , DNA Viral/metabolismo , Farmacorresistência Viral , Endodesoxirribonucleases/genética , Humanos , Mutação de Sentido Incorreto , Proteínas Estruturais Virais/genética
17.
J Antimicrob Chemother ; 65(7): 1347-52, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20453068

RESUMO

OBJECTIVES: Previous studies suggested that helicase-primase inhibitor (HPI) resistance mutations can be selected at relatively high frequency from some isolates of herpes simplex virus type 1 (HSV-1). An intentional mismatch primer (IMP) PCR was developed to detect three known HPI resistance mutations well above the expected background frequency. The objective of this study was to provide proof that HPI resistance mutations pre-exist at relatively high frequency in some clinical isolates obtained from individuals naive to HPIs. METHODS: Three different IMP PCRs were standardized to detect critical HPI resistance mutations (K356N or K356T in UL5, or A899T in UL52) at 10-100 times the expected background frequency (<10(-6)). Thirty HSV-1 clinical isolates were then screened for the resistance mutations in the absence of the inhibitor using IMP PCR. RESULTS: Among 30 clinical isolates that were all susceptible to the HPI, BAY 57-1293, 5 were shown to contain UL5 mutations at 10-100 times higher than the expected frequency. No UL52 resistance mutations were encountered in this study. CONCLUSIONS: The detection of HPI-resistant mutations in some clinical isolates by means of IMP PCR proved that the mutations pre-exist and showed that they are not induced during incubation with the inhibitor.


Assuntos
Antivirais/farmacologia , DNA Primase/genética , Farmacorresistência Viral , Herpes Simples/virologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/genética , Mutação de Sentido Incorreto , Animais , Linhagem Celular , Chlorocebus aethiops , DNA Primase/antagonistas & inibidores , Primers do DNA/genética , DNA Viral/genética , Inibidores Enzimáticos/farmacologia , Herpesvirus Humano 1/isolamento & purificação , Humanos , Reação em Cadeia da Polimerase/métodos , Suínos , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/genética
18.
Antiviral Res ; 87(1): 67-73, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20420855

RESUMO

Point mutations in the HSV-1 UL5 (helicase) gene confer resistance to helicase-primase inhibitors (HPIs), e.g. BAY 57-1293. Such mutations normally occur at a frequency of < or =10(-6)PFU. However, individual HSV-1 laboratory strains and some clinical isolates contained resistance mutations (e.g. UL5: Lys356Asn) at 10(-4)PFU. To address the possibility that pre-existing mutants at high frequency might have an impact on therapy using HPIs, deliberate mixtures were prepared to contain the SC16 UL5: Lys356Asn mutant in SC16 wild-type in the proportion of 1/500 or 1/50PFU. Mice were infected in the neck-skin with 5x10(4)PFU/mouse of wt alone, mutant alone, or the respective mixture. The mutant could not be detected in infectious virus yields from mice inoculated with the 1/500 mixture. However, resistant mutant was recovered from some treated mice inoculated with the 1/50 mixture. All mice inoculated with mixtures remained responsive to BAY 57-1293-therapy with no increase in clinical signs compared to treatment of wt-infected mice.


Assuntos
Antivirais/uso terapêutico , DNA Helicases/antagonistas & inibidores , DNA Primase/antagonistas & inibidores , Farmacorresistência Viral , Inibidores Enzimáticos/uso terapêutico , Herpes Simples/tratamento farmacológico , Herpesvirus Humano 1/efeitos dos fármacos , Proteínas Virais/antagonistas & inibidores , Substituição de Aminoácidos/genética , Animais , DNA Helicases/genética , DNA Primase/genética , Feminino , Herpes Simples/virologia , Herpesvirus Humano 1/enzimologia , Herpesvirus Humano 1/genética , Camundongos , Camundongos Endogâmicos BALB C , Mutação de Sentido Incorreto , Resultado do Tratamento , Proteínas Virais/genética
19.
Antimicrob Agents Chemother ; 54(3): 1290-7, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20047911

RESUMO

Human cytomegalovirus (HCMV) remains a serious threat for immunocompromised individuals, including transplant recipients and newborns. To date, all drugs licensed for the treatment of HCMV infection and disease target the viral DNA polymerase. Although these drugs are effective, several drawbacks are associated with their use, including toxicity and emergence of drug resistance. Hence, new and improved antivirals with novel molecular targets are urgently needed. Here we report on the antiviral properties of AIC246, a representative of a novel class of low-molecular-weight compounds that is currently undergoing clinical phase II studies. The anti-HCMV activity of AIC246 was evaluated in vitro and in vivo using various cell culture assays and an engineered mouse xenograft model. In addition, antiviral properties of the drug were characterized in comparison to the current gold standard ganciclovir. We demonstrate that AIC246 exhibits excellent in vitro inhibitory activity against HCMV laboratory strains and clinical isolates, retains activity against ganciclovir-resistant viruses, is well tolerated in different cell types (median selectivity index, 18,000), and exerts a potent in vivo efficacy in a mouse xenograft model. Moreover, we show that the antiviral block induced by AIC246 is reversible and the efficacy of the drug is not significantly affected by cell culture variations such as cell type or multiplicity of infection. Finally, initial mode-of-action analyses reveal that AIC246 targets a process in the viral replication cycle that occurs later than DNA synthesis. Thus, AIC246 acts via a mode of action that differs from that of polymerase inhibitors like ganciclovir.


Assuntos
Ácido Acético/farmacologia , Antivirais/farmacologia , Infecções por Citomegalovirus/tratamento farmacológico , Citomegalovirus/efeitos dos fármacos , Quinazolinas/farmacologia , Ácido Acético/administração & dosagem , Ácido Acético/química , Animais , Antivirais/administração & dosagem , Antivirais/química , Linhagem Celular , Células Cultivadas , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/virologia , Efeito Citopatogênico Viral , Derme/citologia , Modelos Animais de Doenças , Fibroblastos/virologia , Humanos , Pulmão/citologia , Camundongos , Camundongos SCID , Quinazolinas/administração & dosagem , Quinazolinas/química , Transplante Heterólogo , Resultado do Tratamento , Replicação Viral/efeitos dos fármacos
20.
J Virol ; 83(22): 11902-13, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19740994

RESUMO

The UL84 open reading frame of human cytomegalovirus encodes an essential multifunctional regulatory protein that is thought to act in the nucleus as an initiator of lytic viral replication. Nuclear trafficking of pUL84 is facilitated by a complex nonconventional nuclear localization signal (NLS) that mediates its interaction with the cellular importin-alpha/beta pathway. Since binding of pUL84 to importin-alpha proteins mechanistically differs from that of cellular proteins containing a classical NLS, we assumed that specific interference with the nuclear import of pUL84 might be possible and that this could constitute a novel principle for antiviral therapy. In order to test this hypothesis, we employed peptide aptamer technology and isolated several peptide aptamers from a randomized peptide expression library that specifically bind with high affinity to the unconventional pUL84 NLS under intracellular conditions. Coimmunoprecipitation experiments confirmed these interactions in mammalian cells, and the antiviral potential of the identified peptide aptamers was determined using three independent experimental approaches. (i) Infection experiments with a recombinant human cytomegalovirus expressing green fluorescent protein demonstrated 50 to 60% decreased viral replication in primary human fibroblasts stably expressing pUL84-specific aptamers. (ii) A 50 to 70% reduction of viral plaque formation, as well as a 70 to 90% inhibition of virus release in the presence of pUL84-specific aptamers, was observed. (iii) Immunofluorescence analyses revealed a shift from an almost exclusively nuclear pUL84 staining pattern to a nucleocytoplasmic distribution upon coexpression of the identified molecules, indicating that interference with the nuclear import of pUL84 contributes to the observed antiviral activity of the identified pUL84-binding aptamer molecules.


Assuntos
Aptâmeros de Peptídeos/farmacologia , Citomegalovirus/fisiologia , Transativadores/fisiologia , Replicação Viral/fisiologia , Western Blotting , Linhagem Celular , Fibroblastos/virologia , Técnica Indireta de Fluorescência para Anticorpo , Células HeLa , Humanos , Imunoprecipitação , Ligação Proteica , Transativadores/antagonistas & inibidores , Técnicas do Sistema de Duplo-Híbrido , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA