Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
PLoS Pathog ; 17(3): e1009471, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33780515

RESUMEN

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.


Asunto(s)
Infecciones por Citomegalovirus , Fragmentos Fc de Inmunoglobulinas , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas , Becaplermina/efectos de los fármacos , Becaplermina/metabolismo , Citomegalovirus , Fibroblastos , Células HEK293 , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/farmacología , Mutagénesis Sitio-Dirigida , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/química , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/farmacología
2.
Clin Infect Dis ; 68(8): 1420-1426, 2019 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-30137245

RESUMEN

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.


Asunto(s)
Infecciones por Citomegalovirus/clasificación , Trasplante de Células Madre Hematopoyéticas , Trasplante de Órganos , Receptores de Trasplantes , Farmacorresistencia Viral , Humanos , Huésped Inmunocomprometido , Factores de Riesgo , Terminología como Asunto , Insuficiencia del Tratamiento
3.
J Virol ; 91(12)2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28356534

RESUMEN

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.


Asunto(s)
Citomegalovirus/enzimología , Endodesoxirribonucleasas/metabolismo , Proteínas Virales/metabolismo , Proteínas Estructurales Virales/metabolismo , Transporte Activo de Núcleo Celular/genética , Línea Celular , Cromosomas Artificiales Bacterianos/genética , Citomegalovirus/genética , Citomegalovirus/metabolismo , ADN Viral , Endodesoxirribonucleasas/química , Endodesoxirribonucleasas/genética , Fibroblastos/virología , Genoma Viral , Células HeLa , Humanos , Complejo de la Endopetidasa Proteasomal/genética , Estabilidad Proteica , Proteínas Virales/genética , Proteínas Estructurales Virales/genética
4.
N Engl J Med ; 370(19): 1781-9, 2014 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-24806159

RESUMEN

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.).


Asunto(s)
Acetatos/administración & dosificación , Antivirales/administración & dosificación , Infecciones por Citomegalovirus/prevención & control , Trasplante de Células Madre Hematopoyéticas , Huésped Inmunocomprometido , Infecciones Oportunistas/prevención & control , Quinazolinas/administración & dosificación , Acetatos/efectos adversos , Adulto , Antivirales/efectos adversos , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Humanos , Incidencia , Estimación de Kaplan-Meier , Quinazolinas/efectos adversos , Trasplante Homólogo , Insuficiencia del Tratamiento
5.
J Infect Dis ; 213(1): 23-30, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26113373

RESUMEN

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.


Asunto(s)
Infecciones por Citomegalovirus/tratamiento farmacológico , Infecciones por Citomegalovirus/virología , Citomegalovirus/efectos de los fármacos , Citomegalovirus/genética , Acetatos/farmacología , Acetatos/uso terapéutico , Antivirales/farmacología , Antivirales/uso terapéutico , Infecciones por Citomegalovirus/epidemiología , ADN Viral/sangre , ADN Viral/genética , Farmacorresistencia Viral/efectos de los fármacos , Farmacorresistencia Viral/genética , Humanos , Sistemas de Lectura Abierta/genética , Quinazolinas/farmacología , Quinazolinas/uso terapéutico , Carga Viral , Viremia
6.
Antimicrob Agents Chemother ; 59(6): 3140-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25779572

RESUMEN

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.


Asunto(s)
Acetatos/farmacología , Fármacos Anti-VIH/farmacología , Antivirales/farmacología , Citomegalovirus/efectos de los fármacos , Quinazolinas/farmacología , Línea Celular , Combinación de Medicamentos , Farmacorresistencia Viral , Humanos , Replicación Viral/efectos de los fármacos
7.
Antimicrob Agents Chemother ; 58(1): 610-3, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24189264

RESUMEN

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.


Asunto(s)
Acetatos/farmacología , Antivirales/farmacología , Citomegalovirus/genética , Quinazolinas/farmacología , Citomegalovirus/efectos de los fármacos , ADN Polimerasa Dirigida por ADN/genética , Farmacorresistencia Viral/genética , Humanos
8.
Transpl Int ; 27(1): 77-86, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24164420

RESUMEN

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.


Asunto(s)
Acetatos/administración & dosificación , Infecciones por Citomegalovirus/tratamiento farmacológico , Trasplante de Riñón , Quinazolinas/administración & dosificación , Acetatos/farmacocinética , Adulto , Anciano , Antivirales/administración & dosificación , Antivirales/farmacocinética , Citomegalovirus/genética , Infecciones por Citomegalovirus/virología , ADN Viral/sangre , Femenino , Humanos , Trasplante de Riñón/efectos adversos , Masculino , Persona de Mediana Edad , Infecciones Oportunistas/tratamiento farmacológico , Infecciones Oportunistas/virología , Quinazolinas/farmacocinética , Carga Viral , Viremia/virología
9.
Antimicrob Agents Chemother ; 56(2): 1135-7, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22106211

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Infecciones por Citomegalovirus/virología , Citomegalovirus/efectos de los fármacos , Quinazolinas/farmacología , Antivirales/química , Citomegalovirus/aislamiento & purificación , Farmacorresistencia Viral/efectos de los fármacos , Herpesviridae/clasificación , Herpesviridae/efectos de los fármacos , Humanos , Pruebas de Sensibilidad Microbiana , Quinazolinas/química , Especificidad de la Especie , Virosis/virología , Virus/clasificación , Virus/efectos de los fármacos
10.
J Virol ; 85(20): 10884-93, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21752907

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Citomegalovirus/efectos de los fármacos , Endodesoxirribonucleasas/metabolismo , Proteínas Estructurales Virales/metabolismo , Ensamble de Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Análisis Mutacional de ADN , ADN Viral/metabolismo , Farmacorresistencia Viral , Endodesoxirribonucleasas/genética , Humanos , Mutación Missense , Proteínas Estructurales Virales/genética
11.
Med Microbiol Immunol ; 201(4): 567-79, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22965172

RESUMEN

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.


Asunto(s)
Presentación de Antígeno , Antígenos Virales/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Carioferinas/metabolismo , Fosfoproteínas/inmunología , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas de la Matriz Viral/inmunología , Transporte Activo de Núcleo Celular , Núcleo Celular/química , Células Cultivadas , Citoplasma/química , Humanos , Proteína Exportina 1
12.
Viruses ; 13(3)2021 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-33809234

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Infecciones por Citomegalovirus/virología , Citomegalovirus/metabolismo , Merbromina/farmacología , Proteínas Virales/metabolismo , Virión/metabolismo , Células Cultivadas , Fibroblastos , Humanos , Cultivo Primario de Células , Liberación del Virus , Replicación Viral
13.
Viruses ; 13(3)2021 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-33799898

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Infecciones por Citomegalovirus/tratamiento farmacológico , Citomegalovirus/crecimiento & desarrollo , Proteínas Virales/metabolismo , Liberación del Virus/efectos de los fármacos , Cápside/metabolismo , Proteínas de la Cápside/metabolismo , Línea Celular , Núcleo Celular/virología , Citomegalovirus/efectos de los fármacos , Infecciones por Citomegalovirus/patología , Células HEK293 , Células HeLa , Humanos , Proteínas de la Membrana/metabolismo , Simulación de Dinámica Molecular , Membrana Nuclear/virología , Unión Proteica
14.
Antimicrob Agents Chemother ; 54(3): 1290-7, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20047911

RESUMEN

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.


Asunto(s)
Ácido Acético/farmacología , Antivirales/farmacología , Infecciones por Citomegalovirus/tratamiento farmacológico , Citomegalovirus/efectos de los fármacos , Quinazolinas/farmacología , Ácido Acético/administración & dosificación , Ácido Acético/química , Animales , Antivirales/administración & dosificación , Antivirales/química , Línea Celular , Células Cultivadas , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/virología , Efecto Citopatogénico Viral , Dermis/citología , Modelos Animales de Enfermedad , Fibroblastos/virología , Humanos , Pulmón/citología , Ratones , Ratones SCID , Quinazolinas/administración & dosificación , Quinazolinas/química , Trasplante Heterólogo , Resultado del Tratamiento , Replicación Viral/efectos de los fármacos
15.
J Antimicrob Chemother ; 65(7): 1347-52, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20453068

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , ADN Primasa/genética , Farmacorresistencia Viral , Herpes Simple/virología , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/genética , Mutación Missense , Animales , Línea Celular , Chlorocebus aethiops , ADN Primasa/antagonistas & inhibidores , Cartilla de ADN/genética , ADN Viral/genética , Inhibidores Enzimáticos/farmacología , Herpesvirus Humano 1/aislamiento & purificación , Humanos , Reacción en Cadena de la Polimerasa/métodos , Porcinos , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/genética
16.
J Virol ; 83(22): 11902-13, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19740994

RESUMEN

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.


Asunto(s)
Aptámeros de Péptidos/farmacología , Citomegalovirus/fisiología , Transactivadores/fisiología , Replicación Viral/fisiología , Western Blotting , Línea Celular , Fibroblastos/virología , Técnica del Anticuerpo Fluorescente Indirecta , Células HeLa , Humanos , Inmunoprecipitación , Unión Proteica , Transactivadores/antagonistas & inhibidores , Técnicas del Sistema de Dos Híbridos , Replicación Viral/efectos de los fármacos
17.
Mol Cell Biol ; 26(5): 1631-43, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16478985

RESUMEN

The UL69 gene product of human cytomegalovirus belongs to a family of regulatory proteins conserved among all herpesviruses that have in part been characterized as posttranscriptional transactivators participating in the nuclear export of RNA. Recent experiments suggested that pUL69 also acts as a posttranscriptional activator since it was demonstrated that nucleocytoplasmic shuttling via a CRM1-independent nuclear export signal is a prerequisite for its stimulatory effect on gene expression. Based on these findings we initiated studies to investigate the role of pUL69 in mRNA export and demonstrate that pUL69 efficiently promotes the cytoplasmic accumulation of unspliced RNA. Furthermore, we show that this pUL69 activity is linked to the cellular mRNA export machinery by direct protein interaction with the highly related DEXD/H-box RNA helicases UAP56 and URH49. Particularly, we identified a 12-amino-acid domain within the N terminus of pUL69 which is required for binding to UAP56 and URH49, and we could demonstrate that UAP56 interaction and nucleocytoplasmic shuttling are both prerequisites for pUL69-mediated mRNA export. Thus, we identified a novel cellular target which provides a herpesviral regulatory protein with access to a conserved cellular transport system in order to promote nuclear export of unspliced RNA.


Asunto(s)
Citoplasma/genética , ARN Helicasas/metabolismo , Empalme del ARN , ARN Viral/metabolismo , Transactivadores/metabolismo , Proteínas Virales/metabolismo , Transporte Activo de Núcleo Celular , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células Cultivadas , Citomegalovirus/genética , Citomegalovirus/metabolismo , Citomegalovirus/patogenicidad , Citoplasma/metabolismo , ARN Helicasas DEAD-box , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Células HeLa/virología , Humanos , Carioferinas/genética , Carioferinas/metabolismo , Datos de Secuencia Molecular , Mutación , ARN Helicasas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/genética , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Transactivadores/genética , Proteínas Virales/genética , Proteína Exportina 1
18.
Curr Opin Pharmacol ; 8(5): 541-8, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18662804

RESUMEN

Human cytomegalovirus remains an important pathogen for transplant recipients. To date, a limited number of drugs have been licensed for the treatment of HCMV infection and disease, all sharing the same target molecule, the viral DNA-polymerase. Although combating HCMV with DNA-polymerase inhibitors is effective and has been established for many years, there are several drawbacks associated with the use of these drugs including toxicity and emergence of drug resistance. In order to overcome these problems different treatment options and durations have been assessed and new and improved antiviral drugs with novel molecular targets have been discovered. However, not all of these novel drugs had the properties for success in clinical development, and alternative treatment options with known drugs have been evaluated in parallel. Today the need for an antiviral drug that is potent, safe and well tolerated remains.


Asunto(s)
Antivirales/uso terapéutico , Infecciones por Citomegalovirus/tratamiento farmacológico , Trasplante de Órganos , Animales , Citomegalovirus/efectos de los fármacos , Infecciones por Citomegalovirus/prevención & control , Farmacorresistencia Viral , Humanos
19.
Nucleic Acids Res ; 34(4): 1237-49, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16500893

RESUMEN

The human cytomegalovirus protein pUL69 belongs to a family of regulatory factors that is conserved within the Herpesviridae and includes the proteins ICP27 of herpes simplex virus type 1 and EB2 of Epstein-Barr virus. ICP27 and EB2 have been shown to facilitate the nuclear export of viral mRNAs via interacting with the cellular mRNA export factor REF. Furthermore, direct RNA-binding of these proteins was found to be essential for their stimulating effects on mRNA export. Recently, we demonstrated that pUL69 shares common features with ICP27 and EB2 such as (i) nucleocytoplasmic shuttling and (ii) stimulation of nuclear RNA export via binding to the cellular mRNA export machinery. Here, we demonstrate that pUL69 can also interact with RNA both in vivo and in vitro via a complex N-terminal RNA-binding domain consisting of three arginine-rich motifs. Interestingly, the RNA-binding domain of pUL69 overlaps with both the NLS and the binding site of the cellular mRNA export factors UAP56 and URH49. While the deletion of the UAP56/URH49-binding site abolished pUL69-mediated RNA export, an RNA-binding deficient pUL69 mutant which still interacts with UAP56/URH49 retained its RNA export activity. This surprising finding suggests that, in contrast to its homologues, RNA-binding is not a prerequisite for pUL69-mediated nuclear RNA export.


Asunto(s)
Núcleo Celular/metabolismo , ARN Mensajero/metabolismo , Transactivadores/química , Transactivadores/metabolismo , Proteínas Virales/química , Proteínas Virales/metabolismo , Transporte Activo de Núcleo Celular , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Arginina/análisis , Sitios de Unión , Línea Celular , Humanos , Datos de Secuencia Molecular , Mutación , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Estructura Terciaria de Proteína , Empalme del ARN , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transactivadores/genética , Proteínas Virales/genética
20.
Mol Cell Biol ; 22(16): 5826-34, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12138193

RESUMEN

Sox10 belongs to a family of transcription regulators characterized by a DNA-binding domain known as the HMG box. It plays fundamental roles in neural crest development, peripheral gliogenesis, and terminal differentiation of oligodendrocytes. In accord with its function as transcription factor, Sox10 contains two nuclear localization signals and is most frequently detected in the nucleus. In this study, we report that Sox10 is an active nucleocytoplasmic shuttle protein, competent of both entering and exiting the nucleus. We identified a functional Rev-type nuclear export signal within the DNA-binding domain of Sox10. Mutational inactivation of this nuclear export signal or treatment of cells with the CRM1-specific export inhibitor leptomycin B inhibited nuclear export and consequently nucleocytoplasmic shuttling of Sox10. Importantly, the inhibition of the nuclear export of Sox10 led to decreased transactivation of transfected reporters and endogenous target genes, arguing that continuous nucleocytoplasmic shuttling is essential for the function of Sox10. To our knowledge this is the first time that nuclear export has been reported and shown to be functionally relevant for any Sox protein.


Asunto(s)
Transporte Activo de Núcleo Celular/fisiología , Proteínas de Unión al ADN/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Señales de Clasificación de Proteína/genética , Receptores Citoplasmáticos y Nucleares , Factores de Transcripción/metabolismo , Activación Transcripcional/genética , Animales , Línea Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/genética , Genes Reporteros , Proteínas del Grupo de Alta Movilidad/genética , Humanos , Inmunohistoquímica , Carioferinas/metabolismo , Estructura Secundaria de Proteína , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Transcripción SOXE , Factores de Transcripción/genética , Proteína Exportina 1
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda