Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 273
Filtrar
1.
Sci Rep ; 14(1): 10253, 2024 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-38704431

RESUMEN

The tegument protein pp150 of Human Cytomegalovirus (HCMV) is known to be essential for the final stages of virus maturation and mediates its functions by interacting with capsid proteins. Our laboratory has previously identified the critical regions in pp150 important for pp150-capsid interactions and designed peptides similar in sequence to these regions, with a goal to competitively inhibit capsid maturation. Treatment with a specific peptide (PepCR2 or P10) targeted to pp150 conserved region 2 led to a significant reduction in murine CMV (MCMV) growth in cell culture, paving the way for in vivo testing in a mouse model of CMV infection. However, the general pharmacokinetic parameters of peptides, including rapid degradation and limited tissue and cell membrane permeability, pose a challenge to their successful use in vivo. Therefore, we designed a biopolymer-stabilized elastin-like polypeptide (ELP) fusion construct (ELP-P10) to enhance the bioavailability of P10. Antiviral efficacy and cytotoxic effects of ELP-P10 were studied in cell culture, and pharmacokinetics, biodistribution, and antiviral efficacy were studied in a mouse model of CMV infection. ELP-P10 maintained significant antiviral activity in cell culture, and this conjugation significantly enhanced P10 bioavailability in mouse tissues. The fluorescently labeled ELP-P10 accumulated to higher levels in mouse liver and kidneys as compared to the unconjugated P10. Moreover, viral titers from vital organs of MCMV-infected mice indicated a significant reduction of virus load upon ELP-P10 treatment. Therefore, ELP-P10 has the potential to be developed into an effective antiviral against CMV infection.


Asunto(s)
Antivirales , Infecciones por Citomegalovirus , Elastina , Muromegalovirus , Péptidos , Fosfoproteínas , Proteínas de la Matriz Viral , Animales , Elastina/química , Elastina/metabolismo , Infecciones por Citomegalovirus/tratamiento farmacológico , Infecciones por Citomegalovirus/virología , Ratones , Antivirales/farmacología , Antivirales/farmacocinética , Antivirales/química , Péptidos/farmacología , Péptidos/química , Muromegalovirus/efectos de los fármacos , Humanos , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/química , Citomegalovirus/efectos de los fármacos , Cápside/metabolismo , Cápside/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/farmacocinética , Modelos Animales de Enfermedad , Polipéptidos Similares a Elastina
2.
Eur J Med Chem ; 271: 116402, 2024 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-38636128

RESUMEN

Hepatitis B virus (HBV) capsid assembly modulators (CAMs) represent a promising therapeutic approach for the treatment of HBV infection. In this study, the hit compound CDI (IC50 = 2.46 ± 0.33 µM) was identified by screening of an in-house compound library. And then novel potent benzimidazole derivatives were designed and synthesized as core assembly modulators, and their antiviral effects were evaluated in vitro and in vivo biological experiments. The results indicated that compound 26f displayed the most optimized modulator of HBV capsid assembly (IC50 = 0.51 ± 0.20 µM, EC50 = 2.24 ± 0.43 µM, CC50 = 84.29 µM) and high selectivity index. Moreover, treatment with compound 26f for 14 days significantly decreased serum levels of HBV DNA levels in the Hydrodynamic-Injection (HDI) mouse model. Therefore, compound 26f could be considered as a promising candidate drug for further development of novel HBV CAMs with the desired potency and safety.


Asunto(s)
Antivirales , Bencimidazoles , Virus de la Hepatitis B , Hepatitis B , Bencimidazoles/química , Bencimidazoles/farmacología , Bencimidazoles/síntesis química , Antivirales/farmacología , Antivirales/química , Antivirales/síntesis química , Virus de la Hepatitis B/efectos de los fármacos , Animales , Ratones , Humanos , Hepatitis B/tratamiento farmacológico , Relación Estructura-Actividad , Estructura Molecular , Relación Dosis-Respuesta a Droga , Cápside/efectos de los fármacos , Cápside/metabolismo , Pruebas de Sensibilidad Microbiana , Células Hep G2 , Desarrollo de Medicamentos
3.
J Virol ; 98(3): e0150223, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38315015

RESUMEN

Capsid assembly is critical in the hepatitis B virus (HBV) life cycle, mediated by the viral core protein. Capsid assembly is the target for new anti-viral therapeutics known as capsid assembly modulators (CAMs) of which the CAM-aberrant (CAM-A) class induces aberrant shaped core protein structures and leads to hepatocyte cell death. This study aimed to identify the mechanism of action of CAM-A modulators leading to HBV-infected hepatocyte elimination where CAM-A-mediated hepatitis B surface antigen (HBsAg) reduction was evaluated in a stable HBV replicating cell line and in AAV-HBV-transduced C57BL/6, C57BL/6 SCID, and HBV-infected chimeric mice with humanized livers. Results showed that in vivo treatment with CAM-A modulators induced pronounced reductions in hepatitis B e antigen (HBeAg) and HBsAg, associated with a transient alanine amino transferase (ALT) increase. Both HBsAg and HBeAg reductions and ALT increase were delayed in C57BL/6 SCID and chimeric mice, suggesting that adaptive immune responses may indirectly contribute. However, CD8+ T cell depletion in transduced wild-type mice did not impact antigen reduction, indicating that CD8+ T cell responses are not essential. Transient ALT elevation in AAV-HBV-transduced mice coincided with a transient increase in endoplasmic reticulum stress and apoptosis markers, followed by detection of a proliferation marker. Microarray data revealed antigen presentation pathway (major histocompatibility complex class I molecules) upregulation, overlapping with the apoptosis. Combination treatment with HBV-specific siRNA demonstrated that CAM-A-mediated HBsAg reduction is dependent on de novo core protein translation. To conclude, CAM-A treatment eradicates HBV-infected hepatocytes with high core protein levels through the induction of apoptosis, which can be a promising approach as part of a regimen to achieve functional cure. IMPORTANCE: Treatment with hepatitis B virus (HBV) capsid assembly modulators that induce the formation of aberrant HBV core protein structures (CAM-A) leads to programmed cell death, apoptosis, of HBV-infected hepatocytes and subsequent reduction of HBV antigens, which differentiates CAM-A from other CAMs. The effect is dependent on the de novo synthesis and high levels of core protein.


Asunto(s)
Antivirales , Apoptosis , Regulación Viral de la Expresión Génica , Antígenos del Núcleo de la Hepatitis B , Virus de la Hepatitis B , Hepatocitos , Biosíntesis de Proteínas , Animales , Ratones , Antivirales/farmacología , Antivirales/uso terapéutico , Apoptosis/efectos de los fármacos , Cápside/química , Cápside/clasificación , Cápside/efectos de los fármacos , Cápside/metabolismo , Proteínas de la Cápside/metabolismo , Hepatitis B/tratamiento farmacológico , Hepatitis B/inmunología , Hepatitis B/metabolismo , Hepatitis B/virología , Antígenos del Núcleo de la Hepatitis B/biosíntesis , Antígenos del Núcleo de la Hepatitis B/metabolismo , Antígenos e de la Hepatitis B/metabolismo , Antígenos de Superficie de la Hepatitis B/metabolismo , Virus de la Hepatitis B/crecimiento & desarrollo , Virus de la Hepatitis B/inmunología , Virus de la Hepatitis B/metabolismo , Virus de la Hepatitis B/patogenicidad , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Hepatocitos/virología , Ratones Endogámicos C57BL , Ratones SCID , Replicación Viral , Línea Celular , Linfocitos T CD8-positivos/inmunología , Presentación de Antígeno
4.
J Virol ; 97(10): e0072223, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37754761

RESUMEN

IMPORTANCE: Chronic hepatitis B is the most important cause of liver cancer worldwide and affects more than 290 million people. Current treatments are mostly suppressive and rarely lead to a cure. Therefore, there is a need for novel and curative drugs that target the host or the causative agent, hepatitis B virus itself. Capsid assembly modulators are an interesting class of antiviral molecules that may one day become part of curative treatment regimens for chronic hepatitis B. Here we explore the characteristics of a particularly interesting subclass of capsid assembly modulators. These so-called non-HAP CAM-As have intriguing properties in cell culture but also clear virus-infected cells from the mouse liver in a gradual and sustained way. We believe they represent a considerable improvement over previously reported molecules and may one day be part of curative treatment combinations for chronic hepatitis B.


Asunto(s)
Antivirales , Cápside , Virus de la Hepatitis B , Hepatitis B Crónica , Ensamble de Virus , Animales , Humanos , Ratones , Antivirales/clasificación , Antivirales/farmacología , Antivirales/uso terapéutico , Cápside/química , Cápside/efectos de los fármacos , Cápside/metabolismo , Proteínas de la Cápside/química , Proteínas de la Cápside/efectos de los fármacos , Proteínas de la Cápside/metabolismo , Células Cultivadas , Virus de la Hepatitis B/química , Virus de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/crecimiento & desarrollo , Virus de la Hepatitis B/metabolismo , Hepatitis B Crónica/tratamiento farmacológico , Hepatitis B Crónica/virología , Técnicas In Vitro , Ensamble de Virus/efectos de los fármacos , Modelos Animales de Enfermedad
5.
Bioorg Chem ; 129: 106192, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36265355

RESUMEN

Capsid assembly modulators (CAMs) represent a novel class of antiviral agents targeting hepatitis B virus (HBV) capsid to disrupt the assembly process. NVR 3-778 is the first CAM to demonstrate antiviral activity in patients infected with HBV. However, the relatively low aqueous solubility and moderate activity in the human body halted further development of NVR 3-778. To improve the anti-HBV activity and the drug-like properties of NVR 3-778, we designed and synthesized a series of NVR 3-778 derivatives. Notably, phenylboronic acid-bearing compound 7b (EC50 = 0.83 ± 0.33 µM, CC50 = 19.4 ± 5.0 µM) displayed comparable anti-HBV activity to NVR 3-778 (EC50 = 0.73 ± 0.20 µM, CC50 = 23.4 ± 7.0 µM). Besides, 7b showed improved water solubility (328.8 µg/mL, pH 7) compared to NVR 3-778 (35.8 µg/mL, pH 7). Size exclusion chromatography (SEC) and quantification of encapsidated viral RNA were used to demonstrate that 7b behaves as a class II CAM similar to NVR 3-778. Moreover, molecular dynamics (MD) simulations were conducted to rationalize the structure-activity relationships (SARs) of these novel derivatives and to understand their key interactions with the binding pocket, which provide useful indications for guiding the further rational design of more effective anti-HBV drugs.


Asunto(s)
Antivirales , Benzamidas , Cápside , Diseño de Fármacos , Virus de la Hepatitis B , Ensamble de Virus , Humanos , Antivirales/síntesis química , Antivirales/química , Antivirales/farmacología , Benzamidas/síntesis química , Benzamidas/química , Benzamidas/farmacología , Cápside/efectos de los fármacos , Cápside/metabolismo , Proteínas de la Cápside/metabolismo , Virus de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/fisiología , Ensamble de Virus/efectos de los fármacos
6.
J Virol ; 96(2): e0139521, 2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-34705562

RESUMEN

Viral structural proteins can have multiple activities. Antivirals that target structural proteins have potential to exhibit multiple antiviral mechanisms. Hepatitis B virus (HBV) core protein (Cp) is involved in most stages of the viral life cycle; it assembles into capsids, packages viral RNA, is a metabolic compartment for reverse transcription, interacts with nuclear trafficking machinery, and disassembles to release the viral genome into the nucleus. During nuclear localization, HBV capsids bind to host importins (e.g., Impß) via Cp's C-terminal domain (CTD); the CTD is localized to the interior of the capsid and is transiently exposed on the exterior. We used HAP12 as a representative Cp allosteric modulator (CpAM), a class of antivirals that inappropriately stimulates and misdirects HBV assembly and deforms capsids. CpAM impact on other aspects of the HBV life cycle is poorly understood. We investigate how HAP12 influences the interactions between empty or RNA-filled capsids with Impß and trypsin in vitro. We show that HAP12 can modulate CTD accessibility and capsid stability, depending on the saturation of HAP12-binding sites. We demonstrate that Impß synergistically contributes to capsid disruption at high levels of HAP12 saturation, using electron microscopy to visualize the disruption and rearrangement of Cp dimers into aberrant complexes. However, RNA-filled capsids resist the destabilizing effects of HAP12 and Impß. In summary, we show host protein-induced catalysis of capsid disruption, an unexpected additional mechanism of action for CpAMs. Potentially, untimely capsid disassembly can hamper the HBV life cycle and also cause the virus to become vulnerable to host innate immune responses. IMPORTANCE The HBV core, an icosahedral complex of 120 copies of the homodimeric core (capsid) protein with or without packaged nucleic acid, is transported to the host nucleus by its interaction with host importin proteins. Importin-core interaction requires the core protein C-terminal domain, which is inside the capsid, to "flip" to the capsid exterior. Core protein-directed drugs that affect capsid assembly and stability have been developed recently. We show that these molecules can, synergistically with importins, disrupt capsids. This mechanism of action, synergism with host protein, has the potential to disrupt the virus life cycle and activate the innate immune system.


Asunto(s)
Antivirales/farmacología , Cápside/efectos de los fármacos , Antígenos del Núcleo de la Hepatitis B/química , Virus de la Hepatitis B/efectos de los fármacos , beta Carioferinas/farmacología , Antivirales/química , Cápside/metabolismo , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Antígenos del Núcleo de la Hepatitis B/metabolismo , Unión Proteica , Proteolisis , Ensamble de Virus/efectos de los fármacos , beta Carioferinas/metabolismo
7.
J Virol ; 96(2): e0106021, 2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-34705560

RESUMEN

Rhinoviruses (RVs) cause recurrent infections of the nasal and pulmonary tracts, life-threatening conditions in chronic respiratory illness patients, predisposition of children to asthmatic exacerbation, and large economic cost. RVs are difficult to treat. They rapidly evolve resistance and are genetically diverse. Here, we provide insight into RV drug resistance mechanisms against chemical compounds neutralizing low pH in endolysosomes. Serial passaging of RV-A16 in the presence of the vacuolar proton ATPase inhibitor bafilomycin A1 (BafA1) or the endolysosomotropic agent ammonium chloride (NH4Cl) promoted the emergence of resistant virus populations. We found two reproducible point mutations in viral proteins 1 and 3 (VP1 and VP3), A2526G (serine 66 to asparagine [S66N]), and G2274U (cysteine 220 to phenylalanine [C220F]), respectively. Both mutations conferred cross-resistance to BafA1, NH4Cl, and the protonophore niclosamide, as identified by massive parallel sequencing and reverse genetics, but not the double mutation, which we could not rescue. Both VP1-S66 and VP3-C220 locate at the interprotomeric face, and their mutations increase the sensitivity of virions to low pH, elevated temperature, and soluble intercellular adhesion molecule 1 receptor. These results indicate that the ability of RV to uncoat at low endosomal pH confers virion resistance to extracellular stress. The data endorse endosomal acidification inhibitors as a viable strategy against RVs, especially if inhibitors are directly applied to the airways. IMPORTANCE Rhinoviruses (RVs) are the predominant agents causing the common cold. Anti-RV drugs and vaccines are not available, largely due to rapid evolutionary adaptation of RVs giving rise to resistant mutants and an immense diversity of antigens in more than 160 different RV types. In this study, we obtained insight into the cell biology of RVs by harnessing the ability of RVs to evolve resistance against host-targeting small chemical compounds neutralizing endosomal pH, an important cue for uncoating of normal RVs. We show that RVs grown in cells treated with inhibitors of endolysosomal acidification evolved capsid mutations yielding reduced virion stability against elevated temperature, low pH, and incubation with recombinant soluble receptor fragments. This fitness cost makes it unlikely that RV mutants adapted to neutral pH become prevalent in nature. The data support the concept of host-directed drug development against respiratory viruses in general, notably at low risk of gain-of-function mutations.


Asunto(s)
Cápside/química , Mutación/efectos de los fármacos , Rhinovirus/fisiología , Desencapsidación Viral/fisiología , Antivirales/farmacología , Cápside/efectos de los fármacos , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Farmacorresistencia Viral/efectos de los fármacos , Farmacorresistencia Viral/genética , Endosomas/química , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Molécula 1 de Adhesión Intercelular/metabolismo , Conformación Proteica , Rhinovirus/química , Rhinovirus/efectos de los fármacos , Rhinovirus/genética , Virión/química , Virión/genética , Virión/metabolismo , Internalización del Virus/efectos de los fármacos , Desencapsidación Viral/efectos de los fármacos , Desencapsidación Viral/genética
8.
Nature ; 601(7894): 612-616, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34875675

RESUMEN

Because no currently available vaccine can prevent HIV infection, pre-exposure prophylaxis (PrEP) with antiretrovirals (ARVs) is an important tool for combating the HIV pandemic1,2. Long-acting ARVs promise to build on the success of current PrEP strategies, which must be taken daily, by reducing the frequency of administration3. GS-CA1 is a small-molecule HIV capsid inhibitor with picomolar antiviral potency against a broad array of HIV strains, including variants resistant to existing ARVs, and has shown long-acting therapeutic potential in a mouse model of HIV infection4. Here we show that a single subcutaneous administration of GS-CA1 provides long-term protection against repeated rectal simian-human immunodeficiency virus (SHIV) challenges in rhesus macaques. Whereas all control animals became infected after 15 weekly challenges, a single 300 mg kg-1 dose of GS-CA1 provided per-exposure infection risk reduction of 97% for 24 weeks. Pharmacokinetic analysis showed a correlation between GS-CA1 plasma concentration and protection from SHIV challenges. GS-CA1 levels greater than twice the rhesus plasma protein-adjusted 95% effective concentration conferred 100% protection in this model. These proof-of-concept data support the development of capsid inhibitors as a novel long-acting PrEP strategy in humans.


Asunto(s)
Antirretrovirales , Proteínas de la Cápside , Cápside , Síndrome de Inmunodeficiencia Adquirida del Simio , Virus de la Inmunodeficiencia de los Simios , Animales , Antirretrovirales/farmacología , Cápside/efectos de los fármacos , Proteínas de la Cápside/antagonistas & inhibidores , Proteínas de la Cápside/metabolismo , Macaca mulatta , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/efectos de los fármacos
9.
Front Immunol ; 12: 753467, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34777364

RESUMEN

Adeno-associated viruses (AAV) have emerged as the lead vector in clinical trials and form the basis for several approved gene therapies for human diseases, mainly owing to their ability to sustain robust and long-term in vivo transgene expression, their amenability to genetic engineering of cargo and capsid, as well as their moderate toxicity and immunogenicity. Still, recent reports of fatalities in a clinical trial for a neuromuscular disease, although linked to an exceptionally high vector dose, have raised new caution about the safety of recombinant AAVs. Moreover, concerns linger about the presence of pre-existing anti-AAV antibodies in the human population, which precludes a significant percentage of patients from receiving, and benefitting from, AAV gene therapies. These concerns are exacerbated by observations of cellular immune responses and other adverse events, including detrimental off-target transgene expression in dorsal root ganglia. Here, we provide an update on our knowledge of the immunological and molecular race between AAV (the "hedgehog") and its human host (the "hare"), together with a compendium of state-of-the-art technologies which provide an advantage to AAV and which, thus, promise safer and more broadly applicable AAV gene therapies in the future.


Asunto(s)
Anticuerpos Antivirales/inmunología , Dependovirus/inmunología , Terapia Genética , Vectores Genéticos/inmunología , Inmunidad Adaptativa , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/biosíntesis , Cápside/efectos de los fármacos , Cápside/inmunología , Ensayos Clínicos como Asunto , Dependovirus/clasificación , Dependovirus/genética , Terapia Genética/efectos adversos , Vectores Genéticos/efectos adversos , Humanos , Tolerancia Inmunológica , Inmunidad Celular , Inmunidad Innata , Memoria Inmunológica , Subgrupos Linfocitarios/inmunología , Especificidad de Órganos , Serotipificación , Transgenes
10.
J Virol ; 95(24): e0144521, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34613803

RESUMEN

The HIV-1 capsid, composed of the CA protein, is the target of the novel antiretroviral drug lenacapavir (LCV). CA inhibitors block host factor binding and alter capsid stability to prevent nuclear entry and reverse transcription (RTN), respectively. Capsid stability is mediated in vitro by binding to the host cell metabolite inositol hexakisphosphate (IP6). IP6 depletion in target cells has little effect on HIV-1 infection. We hypothesized that capsid-altering concentrations of CA inhibitors might reveal an effect of IP6 depletion on HIV-1 infection in target cells. To test this, we studied the effects of IP6 depletion on inhibition of infection by the CA inhibitors PF74 and LCV. At low doses of either compound that affect HIV-1 nuclear entry, no effect of IP6 depletion on antiviral activity was observed. Increased antiviral activity was observed in IP6-depleted cells at inhibitor concentrations that affect capsid stability, correlating with increased RTN inhibition. Assays of uncoating and endogenous RTN of purified cores in vitro provided additional support. Our results show that inositol phosphates stabilize the HIV-1 capsid in target cells, thereby dampening the antiviral effects of capsid-targeting antiviral compounds. We propose that targeting of the IP6-binding site in conjunction with CA inhibitors will lead to robust antiretroviral therapy (ART). IMPORTANCE HIV-1 infection and subsequent depletion of CD4+ T cells result in AIDS. Antiretroviral therapy treatment of infected individuals prevents progression to AIDS. The HIV-1 capsid has recently become an ART target. Capsid inhibitors block HIV-1 infection at multiple steps, offering advantages over current ART. The cellular metabolite inositol hexakisphosphate (IP6) binds the HIV-1 capsid, stabilizing it in vitro. However, the function of this interaction in target cells is unclear. Our results imply that IP6 stabilizes the incoming HIV-1 capsid in cells, thus limiting the antiviral efficiency of capsid-destabilizing antivirals. We present a model of capsid inhibitor function and propose that targeting of the IP6-binding site in conjunction with capsid inhibitors currently in development will lead to more robust ART.


Asunto(s)
Fármacos Anti-VIH/farmacología , Cápside/efectos de los fármacos , VIH-1/efectos de los fármacos , Interacciones Huésped-Patógeno/efectos de los fármacos , Fosfatos de Inositol/metabolismo , Ácido Fítico/metabolismo , Proteínas de la Cápside , Línea Celular , Humanos
11.
Molecules ; 26(19)2021 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-34641294

RESUMEN

Supramolecular protein complexes are the corner stone of biological processes; they are essential for many biological functions. Unraveling the interactions responsible for the (dis)assembly of these complexes is required to understand nature and to exploit such systems in future applications. Virus capsids are well-defined assemblies of hundreds of proteins and form the outer shell of non-enveloped viruses. Due to their potential as a drug carriers or nano-reactors and the need for virus inactivation strategies, assessing the intactness of virus capsids is of great interest. Current methods to evaluate the (dis)assembly of these protein assemblies are experimentally demanding in terms of instrumentation, expertise and time. Here we investigate a new strategy to monitor the disassembly of fluorescently labeled virus capsids. To monitor surfactant-induced capsid disassembly, we exploit the complex photophysical interplay between multiple fluorophores conjugated to capsid proteins. The disassembly of the capsid changes the photophysical interactions between the fluorophores, and this can be spectrally monitored. The presented data show that this low complexity method can be used to study and monitor the disassembly of supramolecular protein complexes like virus capsids. However, the range of labeling densities that is suitable for this assay is surprisingly narrow.


Asunto(s)
Cápside/química , Colorantes Fluorescentes/química , Tensoactivos/efectos adversos , Cápside/efectos de los fármacos , Proteínas de la Cápside/química , Proteínas de la Cápside/efectos de los fármacos , Transferencia Resonante de Energía de Fluorescencia , Conformación Proteica , Inactivación de Virus
12.
Viruses ; 13(10)2021 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-34696470

RESUMEN

Feline calicivirus (FCV) causes upper respiratory tract disease (URTD) and sporadic outbreaks of virulent systemic disease (FCV-VSD). The basis for the increased pathogenicity of FCV-VSD viruses is incompletely understood, and antivirals for FCV-VSD have yet to be developed. We investigated the clinicoepidemiology and viral features of three FCV-VSD outbreaks in Australia and evaluated the in vitro efficacy of nitazoxanide (NTZ), 2'-C-methylcytidine (2CMC) and NITD-008 against FCV-VSD viruses. Overall mortality among 23 cases of FCV-VSD was 39%. Metagenomic sequencing identified five genetically distinct FCV lineages within the three outbreaks, all seemingly evolving in situ in Australia. Notably, no mutations that clearly distinguished FCV-URTD from FCV-VSD phenotypes were identified. One FCV-URTD strain likely originated from a recombination event. Analysis of seven amino-acid residues from the hypervariable E region of the capsid in the cultured viruses did not support the contention that properties of these residues can reliably differentiate between the two pathotypes. On plaque reduction assays, dose-response inhibition of FCV-VSD was obtained with all antivirals at low micromolar concentrations; NTZ EC50, 0.4-0.6 µM, TI = 21; 2CMC EC50, 2.7-5.3 µM, TI > 18; NITD-008, 0.5 to 0.9 µM, TI > 111. Investigation of these antivirals for the treatment of FCV-VSD is warranted.


Asunto(s)
Antivirales/uso terapéutico , Calicivirus Felino/aislamiento & purificación , Enfermedades de los Gatos/tratamiento farmacológico , Enfermedades de los Gatos/epidemiología , Brotes de Enfermedades/veterinaria , Animales , Australia , Infecciones por Caliciviridae/patología , Infecciones por Caliciviridae/veterinaria , Infecciones por Caliciviridae/virología , Calicivirus Felino/clasificación , Calicivirus Felino/genética , Cápside/efectos de los fármacos , Enfermedades de los Gatos/patología , Enfermedades de los Gatos/virología , Gatos , Citidina/análogos & derivados , Citidina/uso terapéutico , Femenino , Masculino , Metagenoma , Nitrocompuestos/uso terapéutico , Filogenia , Tiazoles/uso terapéutico
13.
Antiviral Res ; 195: 105177, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34517053

RESUMEN

Vapendavir is a rhino/enterovirus inhibitor that targets a hydrophobic pocket in the viral capsid preventing the virus from entering the cell. We set out to study and compare the molecular mechanisms of resistance to vapendavir among clinically relevant Picornavirus species. To this end in vitro resistance selection of drug-resistant isolates was applied in rhinovirus 2 and 14, enterovirus-D68 and Poliovirus 1 Sabin. Mutations in the drug-binding pocket in VP1 (C199R/Y in hRV14; I194F in PV1; M252L and A156T in EV-D68), typical for this class of compounds, were identified. Interestingly, we also observed mutations located outside the pocket (K167E in EV-D68 and G149C in hRV2) that contribute to the resistant phenotype. Remarkably, the G149C substitution rendered the replication of human rhinovirus 2 dependent on the presence of vapendavir. Our data suggest that the binding of vapendavir to the capsid of the G149C isolate may be required to stabilize the viral particle and to allow efficient dissemination of the virus. We observed the dependency of the G149C isolate on other compounds of this class, suggesting that this phenotype is common for capsid binders. In addition the VP1 region containing the G149C substitution has not been associated with antiviral resistance before. Our results demonstrate that the phenotype and genotype of clinically relevant vapendavir-resistant picornavirus species is more complex than generally believed.


Asunto(s)
Antivirales/farmacología , Farmacorresistencia Viral/genética , Picornaviridae/efectos de los fármacos , Picornaviridae/genética , Replicación Viral/efectos de los fármacos , Animales , Cápside/efectos de los fármacos , Línea Celular , Efecto Citopatogénico Viral , Genotipo , Haplorrinos , Células HeLa , Humanos , Mutación , Fenotipo
14.
Molecules ; 26(13)2021 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-34206893

RESUMEN

PF74 is a capsid-targeting inhibitor of HIV replication that effectively perturbs the highly sensitive viral uncoating process. A lack of information regarding the optical purity (enantiomeric excess) of the single stereogenic centre of PF74 has resulted in ambiguity as to the potency of different samples of this compound. Herein is described the synthesis of enantiomerically enriched (S)- and (R)-PF74 and further enrichment of the samples (≥98%) using chiral HPLC resolution. The biological activities of each enantiomer were then evaluated, which determined (S)-PF74 (IC50 1.5 µM) to be significantly more active than (R)-PF74 (IC50 19 µM). Computational docking studies were then conducted to rationalise this large discrepancy in activity, which indicated different binding conformations for each enantiomer. The binding energy of the conformation adopted by the more active (S)-PF74 (ΔG = -73.8 kcal/mol) was calculated to be more favourable than the conformation adopted by the less active (R)-enantiomer (ΔG = -55.8 kcal/mol) in agreement with experimental observations.


Asunto(s)
Fármacos Anti-VIH/farmacología , Proteínas de la Cápside/metabolismo , Cápside/efectos de los fármacos , Infecciones por VIH/tratamiento farmacológico , VIH-1/efectos de los fármacos , Indoles/farmacología , Fenilalanina/análogos & derivados , Fármacos Anti-VIH/síntesis química , Fármacos Anti-VIH/química , Cápside/química , Cromatografía Líquida de Alta Presión , Células HEK293 , Humanos , Indoles/síntesis química , Indoles/química , Concentración 50 Inhibidora , Simulación del Acoplamiento Molecular , Fenilalanina/síntesis química , Fenilalanina/química , Fenilalanina/farmacología , Estereoisomerismo
15.
Viruses ; 13(5)2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-34063519

RESUMEN

While drug resistance mutations can often be attributed to the loss of direct or solvent-mediated protein-ligand interactions in the drug-mutant complex, in this study we show that a resistance mutation for the picomolar HIV-1 capsid (CA)-targeting antiviral (GS-6207) is mainly due to the free energy cost of the drug-induced protein side chain reorganization in the mutant protein. Among several mutations, M66I causes the most suppression of the GS-6207 antiviral activity (up to ~84,000-fold), and only 83- and 68-fold reductions for PF74 and ZW-1261, respectively. To understand the molecular basis of this drug resistance, we conducted molecular dynamics free energy simulations to study the structures, energetics, and conformational free energy landscapes involved in the inhibitors binding at the interface of two CA monomers. To minimize the protein-ligand steric clash, the I66 side chain in the M66I-GS-6207 complex switches to a higher free energy conformation from the one adopted in the apo M66I. In contrast, the binding of GS-6207 to the wild-type CA does not lead to any significant M66 conformational change. Based on an analysis that decomposes the absolute binding free energy into contributions from two receptor conformational states, it appears that it is the free energy cost of side chain reorganization rather than the reduced protein-ligand interaction that is largely responsible for the drug resistance against GS-6207.


Asunto(s)
Proteínas de la Cápside/genética , Cápside/efectos de los fármacos , Farmacorresistencia Viral/genética , VIH-1/genética , Simulación de Dinámica Molecular , Mutación , Fármacos Anti-VIH/metabolismo , Fármacos Anti-VIH/farmacología , Sitios de Unión , Cápside/química , Cápside/metabolismo , Proteínas de la Cápside/metabolismo , Humanos , Ligandos , Unión Proteica , Conformación Proteica
16.
Viruses ; 13(4)2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33810481

RESUMEN

The hepatitis B virus (HBV) capsid is an attractive drug target, relevant to combating viral hepatitis as a major public health concern. Among small molecules known to interfere with capsid assembly, the phenylpropenamides, including AT130, represent an important antiviral paradigm based on disrupting the timing of genome packaging. Here, all-atom molecular dynamics simulations of an intact AT130-bound HBV capsid reveal that the compound increases spike flexibility and improves recovery of helical secondary structure in the spike tips. Regions of the capsid-incorporated dimer that undergo correlated motion correspond to established sub-domains that pivot around the central chassis. AT130 alters patterns of correlated motion and other essential dynamics. A new conformational state of the dimer is identified, which can lead to dramatic opening of the intradimer interface and disruption of communication within the spike tip. A novel salt bridge is also discovered, which can mediate contact between the spike tip and fulcrum even in closed conformations, revealing a mechanism of direct communication across these sub-domains. Altogether, results describe a dynamical connection between the intra- and interdimer interfaces and enable mapping of allostery traversing the entire core protein dimer.


Asunto(s)
Benzamidas/metabolismo , Proteínas de la Cápside/metabolismo , Cápside/química , Virus de la Hepatitis B/química , Simulación de Dinámica Molecular , Sitio Alostérico , Antivirales/farmacología , Benzamidas/farmacología , Cápside/efectos de los fármacos , Cápside/metabolismo , Biología Computacional/métodos , Virus de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/genética , Humanos , Multimerización de Proteína , Ensamble de Virus
17.
Commun Biol ; 4(1): 250, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33637854

RESUMEN

Enteroviruses pose a persistent and widespread threat to human physical health, with no specific treatments available. Small molecule capsid binders have the potential to be developed as antivirals that prevent virus attachment and entry into host cells. To aid with broad-range drug development, we report here structures of coxsackieviruses B3 and B4 bound to different interprotomer-targeting capsid binders using single-particle cryo-EM. The EM density maps are beyond 3 Å resolution, providing detailed information about interactions in the ligand-binding pocket. Comparative analysis revealed the residues that form a conserved virion-stabilizing network at the interprotomer site, and showed the small molecule properties that allow anchoring in the pocket to inhibit virus disassembly.


Asunto(s)
Antivirales/farmacología , Proteínas de la Cápside/metabolismo , Cápside/efectos de los fármacos , Enterovirus Humano B/efectos de los fármacos , Ensamble de Virus/efectos de los fármacos , Animales , Antivirales/metabolismo , Sitios de Unión , Cápside/metabolismo , Cápside/ultraestructura , Proteínas de la Cápside/ultraestructura , Línea Celular , Chlorocebus aethiops , Microscopía por Crioelectrón , Desarrollo de Medicamentos , Enterovirus Humano B/metabolismo , Enterovirus Humano B/ultraestructura , Ligandos , Simulación del Acoplamiento Molecular , Conformación Proteica
18.
Viruses ; 13(1)2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33467678

RESUMEN

While treatment options are available for hepatitis B virus (HBV), there is currently no cure. Anti-HBV nucleoside analogs and interferon-alpha 2b rarely clear HBV covalently closed circular DNA (cccDNA), requiring lifelong treatment. Recently, we identified GLP-26, a glyoxamide derivative which modulates HBV capsid assembly. The impact of GLP-26 on viral replication and integrated DNA was assessed in an HBV nude mouse model bearing HBV transfected AD38 xenografts. At day 45 post-infection, GLP-26 reduced HBV titers by 2.3-3 log10 versus infected placebo-treated mice. Combination therapy with GLP-26 and entecavir reduced HBV log10 titers by 4.6-fold versus placebo. Next, we examined the pharmacokinetics (PK) in cynomolgus monkeys administered GLP-26 via IV (1 mg/kg) or PO (5 mg/kg). GLP-26 was found to have 34% oral bioavailability, with a mean input time of 3.17 h. The oral dose produced a mean peak plasma concentration of 380.7 ng/mL, observed 0.67 h after administration (~30-fold > in vitro EC90 corrected for protein binding), with a mean terminal elimination half-life of 2.4 h and a mean area under the plasma concentration versus time curve of 1660 ng·hr/mL. GLP-26 was 86.7% bound in monkey plasma. Lastly, GLP-26 demonstrated a favorable toxicity profile confirmed in primary human cardiomyocytes. Thus, GLP-26 warrants further preclinical development as an add on to treatment for HBV infection.


Asunto(s)
Cápside/efectos de los fármacos , Cápside/metabolismo , Cardiotoxinas/farmacocinética , Virus de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/fisiología , Compuestos de Sulfonilurea/farmacocinética , Ensamble de Virus/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Hepatitis B/tratamiento farmacológico , Hepatitis B/virología , Humanos , Macaca fascicularis , Masculino , Ratones , Miocitos Cardíacos/efectos de los fármacos , Compuestos de Sulfonilurea/efectos adversos , Compuestos de Sulfonilurea/química , Carga Viral
19.
Chembiochem ; 22(3): 478-480, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-32856365

RESUMEN

Antivirals are now more important than ever. To efficiently inhibit virus replication, antiviral multivalent strategies need sufficient affinity to overcome the excellent matching between the virus and its receptor. This report highlights a phage capsid scaffold strategy that can be used to precisely position sialic acid moieties to inhibit influenza A virus replication.


Asunto(s)
Antivirales/farmacología , Cápside/efectos de los fármacos , Virus de la Influenza A/efectos de los fármacos , Antivirales/química , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
20.
Sci Rep ; 10(1): 17926, 2020 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-33087754

RESUMEN

Human noroviruses (HuNoVs) are the leading cause of acute gastroenteritis worldwide. Histo-Blood Groups Antigens (HBGAs) have been described as attachment factors, promoting HuNoV infection. However, their role has not yet been elucidated. This study aims to evaluate the ability of HBGAs to protect HuNoVs against various factors naturally found in the human digestive system. The effects of acid pH and proteolytic enzymes (pepsin, trypsin, and chymotrypsin) on GII.4 virus-like particles (VLPs) and GII.4 HuNoVs were studied, both during interactions and non-interaction with HBGAs. The results showed that GII.4 VLPs and GII.4 HuNoVs behaved differently following the treatments. GII.4 VLPs were disrupted at a pH of less than 2.0 and in the presence of proteolytic enzymes (1,500 units/mL pepsin, 100 mg/mL trypsin, and 100 mg/mL chymotrypsin). VLPs were also partially damaged by lower concentrations of trypsin and chymotrypsin (0.1 mg/mL). Conversely, the capsids of GII.4 HuNoVs were not compromised by such treatments, since their genomes were not accessible to RNase. HBGAs were found to offer GII.4 VLPs no protection against an acid pH or proteolytic enzymes.


Asunto(s)
Antígenos de Grupos Sanguíneos/metabolismo , Antígenos de Grupos Sanguíneos/fisiología , Infecciones por Caliciviridae/virología , Gastroenteritis/virología , Norovirus/efectos de los fármacos , Norovirus/patogenicidad , Péptido Hidrolasas/farmacología , Cápside/efectos de los fármacos , Quimotripsina/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Concentración de Iones de Hidrógeno , Norovirus/genética , Norovirus/metabolismo , Pepsina A/farmacología , Tripsina/farmacología , Acoplamiento Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...