Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Cell ; 166(5): 1257-1268.e12, 2016 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-27565348

RESUMEN

While an essential role of HIV-1 integrase (IN) for integration of viral cDNA into human chromosome is established, studies with IN mutants and allosteric IN inhibitors (ALLINIs) have suggested that IN can also influence viral particle maturation. However, it has remained enigmatic as to how IN contributes to virion morphogenesis. Here, we demonstrate that IN directly binds the viral RNA genome in virions. These interactions have specificity, as IN exhibits distinct preference for select viral RNA structural elements. We show that IN substitutions that selectively impair its binding to viral RNA result in eccentric, non-infectious virions without affecting nucleocapsid-RNA interactions. Likewise, ALLINIs impair IN binding to viral RNA in virions of wild-type, but not escape mutant, virus. These results reveal an unexpected biological role of IN binding to the viral RNA genome during virion morphogenesis and elucidate the mode of action of ALLINIs.


Asunto(s)
Genoma Viral , Integrasa de VIH/metabolismo , VIH-1/crecimiento & desarrollo , ARN Viral/metabolismo , Virión/crecimiento & desarrollo , Células HEK293 , Integrasa de VIH/genética , Inhibidores de Integrasa VIH/farmacología , VIH-1/efectos de los fármacos , VIH-1/enzimología , Humanos , Morfogénesis , Nucleocápside/efectos de los fármacos , Unión Proteica , Virión/efectos de los fármacos , Virión/enzimología , Integración Viral/efectos de los fármacos
2.
Langmuir ; 40(2): 1213-1222, 2024 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-38174900

RESUMEN

In biological systems, proteins can bind to nanoparticles to form a "corona" of adsorbed molecules. The nanoparticle corona is of significant interest because it impacts an organism's response to a nanomaterial. Understanding the corona requires knowledge of protein structure, orientation, and dynamics at the surface. A residue-level mapping of protein behavior on nanoparticle surfaces is needed, but this mapping is difficult to obtain with traditional approaches. Here, we have investigated the interaction between R2ab and polystyrene nanoparticles (PSNPs) at the level of individual residues. R2ab is a bacterial surface protein from Staphylococcus epidermidis and is known to interact strongly with polystyrene, leading to biofilm formation. We have used mass spectrometry after lysine methylation and hydrogen-deuterium exchange (HDX) NMR spectroscopy to understand how the R2ab protein interacts with PSNPs of different sizes. Lysine methylation experiments reveal subtle but statistically significant changes in methylation patterns in the presence of PSNPs, indicating altered protein surface accessibility. HDX rates become slower overall in the presence of PSNPs. However, some regions of the R2ab protein exhibit faster than average exchange rates in the presence of PSNPs, while others are slower than the average behavior, suggesting conformational changes upon binding. HDX rates and methylation ratios support a recently proposed "adsorbotope" model for PSNPs, wherein adsorbed proteins consist of unfolded anchor points interspersed with partially structured regions. Our data also highlight the challenges of characterizing complex protein-nanoparticle interactions using these techniques, such as fast exchange rates. While providing insights into how R2ab adsorbs onto PSNP surfaces, this research emphasizes the need for advanced methods to comprehend residue-level interactions in the nanoparticle corona.


Asunto(s)
Nanopartículas , Poliestirenos , Poliestirenos/química , Lisina , Proteínas/química , Nanopartículas/química , Biopelículas
3.
J Biol Chem ; 296: 100363, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33539919

RESUMEN

During the integration step, human immunodeficiency virus type 1 integrase (IN) interacts with viral DNA and the cellular cofactor LEDGF/p75 to effectively integrate the reverse transcript into the host chromatin. Allosteric human immunodeficiency virus type 1 integrase inhibitors (ALLINIs) are a new class of antiviral agents that bind at the dimer interface of the IN catalytic core domain and occupy the binding site of LEDGF/p75. While originally designed to block IN-LEDGF/p75 interactions during viral integration, several of these compounds have been shown to also severely impact viral maturation through an IN multimerization mechanism. In this study, we tested the hypothesis that these dual properties of ALLINIs could be decoupled toward late stage viral replication effects by generating additional contact points between the bound ALLINI and a third subunit of IN. By sequential derivatization at position 7 of a quinoline-based ALLINI scaffold, we show that IN multimerization properties are enhanced by optimizing hydrophobic interactions between the compound and the C-terminal domain of the third IN subunit. These features not only improve the overall antiviral potencies of these compounds but also significantly shift the ALLINIs selectivity toward the viral maturation stage. Thus, we demonstrate that to fully maximize the potency of ALLINIs, the interactions between the inhibitor and all three IN subunits need to be simultaneously optimized.


Asunto(s)
Integrasa de VIH/metabolismo , VIH-1/metabolismo , Quinolinas/farmacología , Regulación Alostérica/efectos de los fármacos , Antivirales/farmacología , Células HEK293 , Integrasa de VIH/fisiología , Inhibidores de Integrasa VIH/metabolismo , Inhibidores de Integrasa VIH/farmacología , VIH-1/efectos de los fármacos , VIH-1/patogenicidad , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Modelos Moleculares , Unión Proteica/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Quinolinas/química , Quinolinas/metabolismo , Integración Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
4.
Molecules ; 23(8)2018 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-30049955

RESUMEN

HIV-1 integrase (IN) inhibitors represent a new class of highly effective anti-AIDS therapeutics. Current FDA-approved IN strand transfer inhibitors (INSTIs) share a common mechanism of action that involves chelation of catalytic divalent metal ions. However, the emergence of IN mutants having reduced sensitivity to these inhibitors underlies efforts to derive agents that antagonize IN function by alternate mechanisms. Integrase along with the 96-residue multifunctional accessory protein, viral protein R (Vpr), are both components of the HIV-1 pre-integration complex (PIC). Coordinated interactions within the PIC are important for viral replication. Herein, we report a 7-mer peptide based on the shortened Vpr (69⁻75) sequence containing a biotin group and a photo-reactive benzoylphenylalanyl residue, and which exhibits low micromolar IN inhibitory potency. Photo-crosslinking experiments have indicated that the peptide directly binds IN. The peptide does not interfere with IN-DNA interactions or induce higher-order, aberrant IN multimerization, suggesting a mode of action for the peptide that is distinct from clinically used INSTIs and developmental allosteric IN inhibitors. This compact Vpr-derived peptide may serve as a valuable pharmacological tool to identify a potential new pharmacologic site.


Asunto(s)
Productos del Gen vpr/química , Productos del Gen vpr/metabolismo , Infecciones por VIH/virología , Inhibidores de Integrasa VIH/farmacología , Integrasa de VIH/metabolismo , VIH-1/fisiología , Péptidos/farmacología , Secuencia de Aminoácidos , Inhibidores de Integrasa VIH/síntesis química , Inhibidores de Integrasa VIH/química , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Péptidos/síntesis química , Péptidos/química , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína
5.
Beilstein J Org Chem ; 14: 2529-2536, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30344776

RESUMEN

A convenient two-step synthesis of ethyl 4-hydroxy-2-methylquinoline-3-carboxylate derivatives has been developed starting from commercially available 2-aminobenzoic acids. In step 1, the anthranilic acids are smoothly converted to isatoic anhydrides using solid triphosgene in THF. In step 2, the anhydride electrophiles are reacted with the sodium enolate of ethyl acetoacetate, generated from sodium hydroxide, in warm N,N-dimethylacetamide resulting in the formation of substituted quinolines. A degradation-build-up strategy of the ethyl ester at the 3-position allowed for the construction of the α-hydroxyacetic acid residue required for the synthesis of key arylquinolines involved in an HIV integrase project.

6.
J Biol Chem ; 291(45): 23569-23577, 2016 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-27645997

RESUMEN

HIV-1 integrase (IN) is essential for virus replication and represents an important multifunctional therapeutic target. Recently discovered quinoline-based allosteric IN inhibitors (ALLINIs) potently impair HIV-1 replication and are currently in clinical trials. ALLINIs exhibit a multimodal mechanism of action by inducing aberrant IN multimerization during virion morphogenesis and by competing with IN for binding to its cognate cellular cofactor LEDGF/p75 during early steps of HIV-1 infection. However, quinoline-based ALLINIs impose a low genetic barrier for the evolution of resistant phenotypes, which highlights a need for discovery of second-generation inhibitors. Using crystallographic screening of a library of 971 fragments against the HIV-1 IN catalytic core domain (CCD) followed by a fragment expansion approach, we have identified thiophenecarboxylic acid derivatives that bind at the CCD-CCD dimer interface at the principal lens epithelium-derived growth factor (LEDGF)/p75 binding pocket. The most active derivative (5) inhibited LEDGF/p75-dependent HIV-1 IN activity in vitro with an IC50 of 72 µm and impaired HIV-1 infection of T cells at an EC50 of 36 µm The identified lead compound, with a relatively small molecular weight (221 Da), provides an optimal building block for developing a new class of inhibitors. Furthermore, although structurally distinct thiophenecarboxylic acid derivatives target a similar pocket at the IN dimer interface as the quinoline-based ALLINIs, the lead compound, 5, inhibited IN mutants that confer resistance to quinoline-based compounds. Collectively, our findings provide a plausible path for structure-based development of second-generation ALLINIs.


Asunto(s)
Infecciones por VIH/tratamiento farmacológico , Inhibidores de Integrasa VIH/química , Inhibidores de Integrasa VIH/farmacología , Integrasa de VIH/metabolismo , VIH-1/efectos de los fármacos , Tiofenos/química , Tiofenos/farmacología , Regulación Alostérica/efectos de los fármacos , Ácidos Carboxílicos/química , Ácidos Carboxílicos/farmacología , Dominio Catalítico/efectos de los fármacos , Cristalografía por Rayos X , Descubrimiento de Drogas , Células HEK293 , Infecciones por VIH/virología , Integrasa de VIH/química , Humanos , Modelos Moleculares , Simulación del Acoplamiento Molecular
7.
PLoS Pathog ; 10(5): e1004171, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24874515

RESUMEN

The quinoline-based allosteric HIV-1 integrase (IN) inhibitors (ALLINIs) are promising candidates for clinically useful antiviral agents. Studies using these compounds have highlighted the role of IN in both early and late stages of virus replication. However, dissecting the exact mechanism of action of the quinoline-based ALLINIs has been complicated by the multifunctional nature of these inhibitors because they both inhibit IN binding with its cofactor LEDGF/p75 and promote aberrant IN multimerization with similar potencies in vitro. Here we report design of small molecules that allowed us to probe the role of HIV-1 IN multimerization independently from IN-LEDGF/p75 interactions in infected cells. We altered the rigid quinoline moiety in ALLINIs and designed pyridine-based molecules with a rotatable single bond to allow these compounds to bridge between interacting IN subunits optimally and promote oligomerization. The most potent pyridine-based inhibitor, KF116, potently (EC50 of 0.024 µM) blocked HIV-1 replication by inducing aberrant IN multimerization in virus particles, whereas it was not effective when added to target cells. Furthermore, KF116 inhibited the HIV-1 IN variant with the A128T substitution, which confers resistance to the majority of quinoline-based ALLINIs. A genome-wide HIV-1 integration site analysis demonstrated that addition of KF116 to target or producer cells did not affect LEDGF/p75-dependent HIV-1 integration in host chromosomes, indicating that this compound is not detectably inhibiting IN-LEDGF/p75 binding. These findings delineate the significance of correctly ordered IN structure for HIV-1 particle morphogenesis and demonstrate feasibility of exploiting IN multimerization as a therapeutic target. Furthermore, pyridine-based compounds present a novel class of multimerization selective IN inhibitors as investigational probes for HIV-1 molecular biology.


Asunto(s)
Antivirales/farmacología , Inhibidores de Integrasa VIH/farmacología , Integrasa de VIH/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Multimerización de Proteína/efectos de los fármacos , Integración Viral/efectos de los fármacos , Línea Celular , VIH-1/efectos de los fármacos , Humanos , Unión Proteica/efectos de los fármacos , Quinolinas/farmacología , Replicación Viral/efectos de los fármacos
8.
Curr Top Microbiol Immunol ; 389: 93-119, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25778682

RESUMEN

Multimeric HIV-1 integrase (IN) plays an essential, multifunctional role in virus replication and serves as an important therapeutic target. Structural and biochemical studies have revealed the importance of the ordered interplay between IN molecules for its function. In the presence of viral DNA ends, individual IN subunits assemble into a tetramer and form a stable synaptic complex (SSC), which mediates integration of the reverse transcribed HIV-1 genome into chromatin. Cellular chromatin-associated protein LEDGF/p75 engages the IN tetramer in the SSC and directs HIV-1 integration into active genes. A mechanism to deregulate the productive interplay between IN subunits with small molecule inhibitors has recently received considerable attention. Most notably, allosteric IN inhibitors (ALLINIs) have been shown to bind to the IN dimer interface at the LEDGF/p75 binding pocket, stabilize interacting IN subunits, and promote aberrant, higher order IN multimerization. Consequently, these compounds impair formation of the SSC and associated LEDGF/p75-independent IN catalytic activities as well as inhibit LEDGF/p75 binding to the SSC in vitro. However, in infected cells, ALLINIs more potently impaired correct maturation of virus particles than the integration step. ALLINI treatments induced aberrant, higher order IN multimerization in virions and resulted in eccentric, non-infectious virus particles. These studies have suggested that the correctly ordered IN structure is important for virus particle morphogenesis and highlighted IN multimerization as a plausible therapeutic target for developing new inhibitors to enhance treatment options for HIV-1-infected patients.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida/tratamiento farmacológico , Inhibidores de Integrasa VIH/uso terapéutico , Integrasa de VIH/fisiología , Multimerización de Proteína/efectos de los fármacos , Integrasa de VIH/química , Humanos , Subunidades de Proteína
9.
Bioorg Med Chem Lett ; 26(19): 4748-4752, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27568085

RESUMEN

Employing a scaffold hopping approach, a series of allosteric HIV-1 integrase (IN) inhibitors (ALLINIs) have been synthesized based on an indole scaffold. These compounds incorporate the key elements utilized in quinoline-based ALLINIs for binding to the IN dimer interface at the principal LEDGF/p75 binding pocket. The most potent of these compounds displayed good activity in the LEDGF/p75 dependent integration assay (IC50=4.5µM) and, as predicted based on the geometry of the five- versus six-membered ring, retained activity against the A128T IN mutant that confers resistance to many quinoline-based ALLINIs.


Asunto(s)
Inhibidores de Integrasa VIH/farmacología , Integrasa de VIH/efectos de los fármacos , Indoles/farmacología , Regulación Alostérica , Cristalografía por Rayos X , Inhibidores de Integrasa VIH/química , Enlace de Hidrógeno , Relación Estructura-Actividad
10.
Proc Natl Acad Sci U S A ; 110(29): 12036-41, 2013 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-23818621

RESUMEN

The selection of chromosomal targets for retroviral integration varies markedly, tracking with the genus of the retrovirus, suggestive of targeting by binding to cellular factors. γ-Retroviral murine leukemia virus (MLV) DNA integration into the host genome is favored at transcription start sites, but the underlying mechanism for this preference is unknown. Here, we have identified bromodomain and extraterminal domain (BET) proteins (Brd2, -3, -4) as cellular-binding partners of MLV integrase. We show that purified recombinant Brd4(1-720) binds with high affinity to MLV integrase and stimulates correct concerted integration in vitro. JQ-1, a small molecule that selectively inhibits interactions of BET proteins with modified histone sites impaired MLV but not HIV-1 integration in infected cells. Comparison of the distribution of BET protein-binding sites analyzed using ChIP-Seq data and MLV-integration sites revealed significant positive correlations. Antagonism of BET proteins, via JQ-1 treatment or RNA interference, reduced MLV-integration frequencies at transcription start sites. These findings elucidate the importance of BET proteins for MLV integration efficiency and targeting and provide a route to developing safer MLV-based vectors for human gene therapy.


Asunto(s)
Integrasas/metabolismo , Virus de la Leucemia Murina/enzimología , Proteínas Nucleares/metabolismo , Proteínas Recombinantes/metabolismo , Factores de Transcripción/metabolismo , Sitio de Iniciación de la Transcripción/fisiología , Integración Viral/fisiología , Animales , Azepinas , Proteínas de Ciclo Celular , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Células HEK293 , Secuenciación de Nucleótidos de Alto Rendimiento , Interacciones Huésped-Patógeno , Humanos , Espectrometría de Masas , Ratones , Células 3T3 NIH , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteómica/métodos , Interferencia de ARN , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Triazoles , Integración Viral/genética
11.
Proc Natl Acad Sci U S A ; 110(21): 8690-5, 2013 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-23610442

RESUMEN

Integration is essential for HIV-1 replication, and the viral integrase (IN) protein is an important therapeutic target. Allosteric IN inhibitors (ALLINIs) that engage the IN dimer interface at the binding site for the host protein lens epithelium-derived growth factor (LEDGF)/transcriptional coactivator p75 are an emerging class of small molecule antagonists. Consistent with the inhibition of a multivalent drug target, ALLINIs display steep antiviral dose-response curves ex vivo. ALLINIs multimerize IN protein and concordantly block its assembly with viral DNA in vitro, indicating that the disruption of two integration-associated functions, IN catalysis and the IN-LEDGF/p75 interaction, determines the multimode mechanism of ALLINI action. We now demonstrate that ALLINI potency is unexpectedly accounted for during the late phase of HIV-1 replication. The compounds promote virion IN multimerization and, reminiscent of class II IN mutations, block the formation of the electron-dense viral core and inhibit reverse transcription and integration in subsequently infected target cells. Mature virions are recalcitrant to ALLINI treatment, and compound potency during virus production is independent of the level of LEDGF/p75 expression. We conclude that cooperative multimerization of IN by ALLINIs together with the inability for LEDGF/p75 to effectively engage the virus during its egress from cells underscores the multimodal mechanism of ALLINI action. Our results highlight the versatile nature of allosteric inhibitors to primarily inhibit viral replication at a step that is distinct from the catalytic requirement for the target enzyme. The vulnerability of IN to small molecules during the late phase of HIV-1 replication unveils a pharmacological Achilles' heel for exploitation in clinical ALLINI development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Inhibidores de Integrasa VIH , Integrasa de VIH/metabolismo , VIH-1/fisiología , Factores de Transcripción/metabolismo , Replicación Viral/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/genética , Regulación Alostérica/efectos de los fármacos , Línea Celular , Inhibidores de Integrasa VIH/química , Inhibidores de Integrasa VIH/farmacología , Humanos , Factores de Transcripción/genética , Virión/genética , Virión/metabolismo , Replicación Viral/fisiología
12.
J Biol Chem ; 289(38): 26430-26440, 2014 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-25118283

RESUMEN

Allosteric HIV-1 integrase (IN) inhibitors (ALLINIs) are a promising class of antiretroviral agents for clinical development. Although ALLINIs promote aberrant IN multimerization and inhibit IN interaction with its cellular cofactor LEDGF/p75 with comparable potencies in vitro, their primary mechanism of action in infected cells is through inducing aberrant multimerization of IN. Crystal structures have shown that ALLINIs bind at the IN catalytic core domain dimer interface and bridge two interacting subunits. However, how these interactions promote higher-order protein multimerization is not clear. Here, we used mass spectrometry-based protein footprinting to monitor surface topology changes in full-length WT and the drug-resistant A128T mutant INs in the presence of ALLINI-2. These experiments have identified protein-protein interactions that extend beyond the direct inhibitor binding site and which lead to aberrant multimerization of WT but not A128T IN. Specifically, we demonstrate that C-terminal residues Lys-264 and Lys-266 play an important role in the inhibitor induced aberrant multimerization of the WT protein. Our findings provide structural clues for exploiting IN multimerization as a new, attractive therapeutic target and are expected to facilitate development of improved inhibitors.


Asunto(s)
Acetatos/química , Inhibidores de Integrasa VIH/química , Integrasa de VIH/química , Indoles/química , Regulación Alostérica , Farmacorresistencia Viral , Integrasa de VIH/genética , VIH-1/enzimología , Humanos , Mutación Missense , Unión Proteica , Multimerización de Proteína/efectos de los fármacos
13.
J Biol Chem ; 288(22): 15813-20, 2013 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-23615903

RESUMEN

Allosteric HIV-1 integrase (IN) inhibitors (ALLINIs) are a very promising new class of anti-HIV-1 agents that exhibit a multimodal mechanism of action by allosterically modulating IN multimerization and interfering with IN-lens epithelium-derived growth factor (LEDGF)/p75 binding. Selection of viral strains under ALLINI pressure has revealed an A128T substitution in HIV-1 IN as a primary mechanism of resistance. Here, we elucidated the structural and mechanistic basis for this resistance. The A128T substitution did not affect the hydrogen bonding between ALLINI and IN that mimics the IN-LEDGF/p75 interaction but instead altered the positioning of the inhibitor at the IN dimer interface. Consequently, the A128T substitution had only a minor effect on the ALLINI IC50 values for IN-LEDGF/p75 binding. Instead, ALLINIs markedly altered the multimerization of IN by promoting aberrant higher order WT (but not A128T) IN oligomers. Accordingly, WT IN catalytic activities and HIV-1 replication were potently inhibited by ALLINIs, whereas the A128T substitution in IN resulted in significant resistance to the inhibitors both in vitro and in cell culture assays. The differential multimerization of WT and A128T INs induced by ALLINIs correlated with the differences in infectivity of HIV-1 progeny virions. We conclude that ALLINIs primarily target IN multimerization rather than IN-LEDGF/p75 binding. Our findings provide the structural foundations for developing improved ALLINIs with increased potency and decreased potential to select for drug resistance.


Asunto(s)
Farmacorresistencia Viral/efectos de los fármacos , Inhibidores de Integrasa VIH/farmacología , Integrasa de VIH/metabolismo , VIH-1/fisiología , Mutación Missense , Multimerización de Proteína/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/genética , Sustitución de Aminoácidos , Células HEK293 , Integrasa de VIH/química , Integrasa de VIH/genética , Inhibidores de Integrasa VIH/química , VIH-1/química , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Unión Proteica
14.
Retrovirology ; 11: 100, 2014 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-25421939

RESUMEN

BACKGROUND: Allosteric HIV-1 integrase (IN) inhibitors (ALLINIs) are an important new class of anti-HIV-1 agents. ALLINIs bind at the IN catalytic core domain (CCD) dimer interface occupying the principal binding pocket of its cellular cofactor LEDGF/p75. Consequently, ALLINIs inhibit HIV-1 IN interaction with LEDGF/p75 as well as promote aberrant IN multimerization. Selection of viral strains emerging under the inhibitor pressure has revealed mutations at the IN dimer interface near the inhibitor binding site. RESULTS: We have investigated the effects of one of the most prevalent substitutions, H171T IN, selected under increasing pressure of ALLINI BI-D. Virus containing the H171T IN substitution exhibited an ~68-fold resistance to BI-D treatment in infected cells. These results correlated with ~84-fold reduced affinity for BI-D binding to recombinant H171T IN CCD protein compared to its wild type (WT) counterpart. However, the H171T IN substitution only modestly affected IN-LEDGF/p75 binding and allowed HIV-1 containing this substitution to replicate at near WT levels. The x-ray crystal structures of BI-D binding to WT and H171T IN CCD dimers coupled with binding free energy calculations revealed the importance of the Nδ- protonated imidazole group of His171 for hydrogen bonding to the BI-D tert-butoxy ether oxygen and establishing electrostatic interactions with the inhibitor carboxylic acid, whereas these interactions were compromised upon substitution to Thr171. CONCLUSIONS: Our findings reveal a distinct mechanism of resistance for the H171T IN mutation to ALLINI BI-D and indicate a previously undescribed role of the His171 side chain for binding the inhibitor.


Asunto(s)
Acetatos/metabolismo , Farmacorresistencia Viral , Inhibidores de Integrasa VIH/metabolismo , Integrasa de VIH/metabolismo , VIH-1/efectos de los fármacos , VIH-1/enzimología , Mutación Missense , Quinolinas/metabolismo , Línea Celular , Cristalografía por Rayos X , Integrasa de VIH/química , Integrasa de VIH/genética , Histidina/genética , Histidina/metabolismo , Humanos , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Unión Proteica , Conformación Proteica
15.
Viruses ; 16(2)2024 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-38399977

RESUMEN

Allosteric HIV-1 Integrase (IN) Inhibitors or ALLINIs bind at the dimer interface of the IN, away from the enzymatic catalytic site, and disable viral replication by inducing over-multimerization of IN. Interestingly, these inhibitors are capable of impacting both the early and late stages of viral replication. To better understand the important binding features of multi-substituted quinoline-based ALLINIs, we have surveyed published studies on IN multimerization and antiviral properties of various substituted quinolines at the 4, 6, 7, and 8 positions. Here we show how the efficacy of these inhibitors can be modulated by the nature of the substitutions at those positions. These features not only improve the overall antiviral potencies of these compounds but also significantly shift the selectivity toward the viral maturation stage. Thus, to fully maximize the potency of ALLINIs, the interactions between the inhibitor and multiple IN subunits need to be simultaneously optimized.


Asunto(s)
Inhibidores de Integrasa VIH , Integrasa de VIH , VIH-1 , Quinolinas , VIH-1/metabolismo , Regulación Alostérica , Inhibidores de Integrasa VIH/farmacología , Inhibidores de Integrasa VIH/química , Integrasa de VIH/metabolismo , Quinolinas/farmacología , Multimerización de Proteína
16.
J Biol Chem ; 287(20): 16801-11, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22437836

RESUMEN

The multifunctional HIV-1 enzyme integrase interacts with viral DNA and its key cellular cofactor LEDGF to effectively integrate the reverse transcript into a host cell chromosome. These interactions are crucial for HIV-1 replication and present attractive targets for antiviral therapy. Recently, 2-(quinolin-3-yl) acetic acid derivatives were reported to selectively inhibit the integrase-LEDGF interaction in vitro and impair HIV-1 replication in infected cells. Here, we show that this class of compounds impairs both integrase-LEDGF binding and LEDGF-independent integrase catalytic activities with similar IC(50) values, defining them as bona fide allosteric inhibitors of integrase function. Furthermore, we show that 2-(quinolin-3-yl) acetic acid derivatives block the formation of the stable synaptic complex between integrase and viral DNA by allosterically stabilizing an inactive multimeric form of integrase. In addition, these compounds inhibit LEDGF binding to the stable synaptic complex. This multimode mechanism of action concordantly results in cooperative inhibition of the concerted integration of viral DNA ends in vitro and HIV-1 replication in cell culture. Our findings, coupled with the fact that high cooperativity of antiviral inhibitors correlates with their increased instantaneous inhibitory potential, an important clinical parameter, argue strongly that improved 2-(quinolin-3-yl) acetic acid derivatives could exhibit desirable clinical properties.


Asunto(s)
ADN Viral/metabolismo , Integrasa de VIH/metabolismo , VIH-1/fisiología , Inhibidores de Integrasa/farmacología , Replicación Viral/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/genética , ADN Viral/genética , Células HEK293 , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/enzimología , Infecciones por VIH/genética , Integrasa de VIH/genética , Humanos , Ácidos Indolacéticos/química , Ácidos Indolacéticos/farmacología , Inhibidores de Integrasa/química , Unión Proteica/efectos de los fármacos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Replicación Viral/fisiología
17.
J Biol Chem ; 287(41): 34044-58, 2012 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-22872640

RESUMEN

Transportin 3 (TNPO3 or TRN-SR2) has been shown to be an important cellular factor for early steps of lentiviral replication. However, separate studies have implicated distinct mechanisms for TNPO3 either through its interaction with HIV-1 integrase or capsid. Here we have carried out a detailed biophysical characterization of TNPO3 and investigated its interactions with viral proteins. Biophysical analyses including circular dichroism, analytical ultracentrifugation, small-angle x-ray scattering, and homology modeling provide insight into TNPO3 architecture and indicate that it is highly structured and exists in a monomer-dimer equilibrium in solution. In vitro biochemical binding assays argued against meaningful direct interaction between TNPO3 and the capsid cores. Instead, TNPO3 effectively bound to the functional intasome but not to naked viral DNA, suggesting that TNPO3 can directly engage the HIV-1 IN tetramer prebound to the cognate DNA. Mass spectrometry-based protein footprinting and site-directed mutagenesis studies have enabled us to map several interacting amino acids in the HIV-1 IN C-terminal domain and the cargo binding domain of TNPO3. Our findings provide important information for future genetic analysis to better understand the role of TNPO3 and its interacting partners for HIV-1 replication.


Asunto(s)
ADN Viral/química , VIH-1/química , beta Carioferinas/química , Dicroismo Circular , ADN Viral/genética , ADN Viral/metabolismo , Integrasa de VIH , VIH-1/fisiología , Humanos , Espectrometría de Masas , Unión Proteica , Estructura Terciaria de Proteína , Dispersión del Ángulo Pequeño , Replicación Viral/fisiología , Difracción de Rayos X , beta Carioferinas/genética , beta Carioferinas/metabolismo
18.
Nucleic Acids Res ; 39(20): 9009-22, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21771857

RESUMEN

A tetramer of HIV-1 integrase (IN) stably associates with the viral DNA ends to form a fully functional concerted integration intermediate. LEDGF/p75, a key cellular binding partner of the lentiviral enzyme, also stabilizes a tetrameric form of IN. However, functional assays have indicated the importance of the order of viral DNA and LEDGF/p75 addition to IN for productive concerted integration. Here, we employed Förster Resonance Energy Transfer (FRET) to monitor assembly of individual IN subunits into tetramers in the presence of viral DNA and LEDGF/p75. The IN-viral DNA and IN-LEDGF/p75 complexes yielded significantly different FRET values suggesting two distinct IN conformations in these complexes. Furthermore, the order of addition experiments indicated that FRET for the preformed IN-viral DNA complex remained unchanged upon its subsequent binding to LEDGF/p75, whereas pre-incubation of LEDGF/p75 and IN followed by addition of viral DNA yielded FRET very similar to the IN-LEDGF/p75 complex. These findings provide new insights into the structural organization of IN subunits in functional concerted integration intermediates and suggest that differential multimerization of IN in the presence of various ligands could be exploited as a plausible therapeutic target for development of allosteric inhibitors.


Asunto(s)
ADN Viral/química , Integrasa de VIH/química , Péptidos y Proteínas de Señalización Intercelular/química , ADN Viral/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Integrasa de VIH/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Modelos Moleculares , Conformación Proteica , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo
19.
bioRxiv ; 2023 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-37693402

RESUMEN

In biological systems, proteins can bind to nanoparticles to form a "corona" of adsorbed molecules. The nanoparticle corona is of high interest because it impacts the organism's response to the nanomaterial. Understanding the corona requires knowledge of protein structure, orientation, and dynamics at the surface. Ultimately, a residue-level mapping of protein behavior on nanoparticle surfaces is needed, but this mapping is difficult to obtain with traditional approaches. Here, we have investigated the interaction between R2ab and polystyrene nanoparticles (PSNPs) at the level of individual residues. R2ab is a bacterial surface protein from Staphylococcus epidermidis and is known to interact strongly with polystyrene, leading to biofilm formation. We have used mass spectrometry after lysine methylation and hydrogen-deuterium exchange (HDX) NMR spectroscopy to understand how the R2ab protein interacts with PSNPs of different sizes. Through lysine methylation, we observe subtle but statistically significant changes in methylation patterns in the presence of PSNPs, indicating altered protein surface accessibility. HDX measurements reveal that certain regions of the R2ab protein undergo faster exchange rates in the presence of PSNPs, suggesting conformational changes upon binding. Both results support a recently proposed "adsorbotope" model, wherein adsorbed proteins consist of unfolded anchor points interspersed with regions of partial structure. Our data also highlight the challenges of characterizing complex protein-nanoparticle interactions using these techniques, such as fast exchange rates. While providing insights into how proteins respond to nanoparticle surfaces, this research emphasizes the need for advanced methods to comprehend these intricate interactions fully at the residue level.

20.
Viruses ; 14(7)2022 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-35891446

RESUMEN

Allosteric HIV-1 integrase (IN) inhibitors, or ALLINIs, are a new class of antiviral agents that bind at the dimer interface of the IN, away from the enzymatic catalytic site and block viral replication by triggering an aberrant multimerization of the viral enzyme. To further our understanding of the important binding features of multi-substituted quinoline-based ALLINIs, we have examined the IN multimerization and antiviral properties of substitution patterns at the 6 or 8 position. We found that the binding properties of these ALLINIs are negatively impacted by the presence of bulky substitutions at these positions. In addition, we have observed that the addition of bromine at either the 6 (6-bromo) or 8 (8-bromo) position conferred better antiviral properties. Finally, we found a significant loss of potency with the 6-bromo when tested with the ALLINI-resistant IN A128T mutant virus, while the 8-bromo analog retained full effectiveness.


Asunto(s)
Inhibidores de Integrasa VIH , Integrasa de VIH , VIH-1 , Quinolinas , Regulación Alostérica , Antivirales/farmacología , Integrasa de VIH/metabolismo , Inhibidores de Integrasa VIH/química , Inhibidores de Integrasa VIH/farmacología , VIH-1/metabolismo , Quinolinas/farmacología , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA