Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Virol ; 93(20)2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31375580

RESUMO

Long-acting antiretrovirals could provide a useful alternative to daily oral therapy for HIV-1-infected individuals. Building on a bi-specific molecule with adnectins targeting CD4 and gp41, a potential long-acting biologic, GSK3732394, was developed with three independent and synergistic modes of HIV entry inhibition that potentially could be self-administered as a long-acting subcutaneous injection. Starting with the bi-specific inhibitor, an α-helical peptide inhibitor was optimized as a linked molecule to the anti-gp41 adnectin, with each separate inhibitor exhibiting at least single-digit nanomolar (or lower) potency and a broad spectrum. Combination of the two adnectins and peptide activities into a single molecule was shown to have synergistic advantages in potency, the resistance barrier, and the ability to inhibit HIV-1 infections at low levels of CD4 receptor occupancy, showing that GSK3732394 can work in trans on a CD4+ T cell. Addition of a human serum albumin molecule prolongs the half-life in a human CD4 transgenic mouse, suggesting that it may have potential as a long-acting agent. GSK3732394 was shown to be highly effective in a humanized mouse model of infection. GSK3732394 is currently in clinical trials.IMPORTANCE There continue to be significant unmet medical needs for patients with HIV-1 infection. One way to improve adherence and decrease the likelihood of drug-drug interactions in HIV-1-infected patients is through the development of long-acting biologic inhibitors. Building on a bi-specific inhibitor approach targeting CD4 and gp41, a tri-specific molecule was generated with three distinct antiviral activities. The linkage of these three biologic inhibitors creates synergy that offers a series of advantages to the molecule. The addition of human serum albumin to the tri-specific inhibitor could allow it to function as a long-acting self-administered treatment for patients with HIV infection. This molecule is currently in early clinical trials.


Assuntos
Inibidores da Fusão de HIV/farmacologia , HIV-1/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Modelos Animais de Doenças , Farmacorresistência Viral , Inibidores da Fusão de HIV/química , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , Humanos , Camundongos , Camundongos Transgênicos , Modelos Moleculares , Peptídeos/química , Peptídeos/farmacologia , Conformação Proteica
2.
J Virol ; 92(14)2018 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-29743355

RESUMO

The N17 region of gp41 in HIV-1 is the most conserved region in gp160. mRNA selection technologies were used to identify an adnectin that binds to this region and inhibits gp41-induced membrane fusion. Additional selection conditions were used to optimize the adnectin to greater potency (5.4 ± 2.6 nM) against HIV-1 and improved binding affinity for an N17-containing helical trimer (0.8 ± 0.4 nM). Resistance to this adnectin mapped to a single Glu-to-Arg change within the N17 coding region. The optimized adnectin (6200_A08) exhibited high potency and broad-spectrum activity against 123 envelope proteins and multiple clinical virus isolates, although certain envelope proteins did exhibit reduced susceptibility to 6200_A08 alone. The reduced potency could not be correlated with sequence changes in the target region and was thought to be the result of faster kinetics of fusion mediated by these envelope proteins. Optimized linkage of 6200_A08 with a previously characterized adnectin targeting CD4 produced a highly synergistic molecule, with the potency of the tandem molecule measured at 37 ± 1 pM. In addition, these tandem molecules now exhibited few potency differences against the same panel of envelope proteins with reduced susceptibility to 6200_A08 alone, providing evidence that they did not have intrinsic resistance to 6200_A08 and that coupling 6200_A08 with the anti-CD4 adnectin may provide a higher effective on rate for gp41 target engagement.IMPORTANCE There continue to be significant unmet medical needs for patients with HIV-1 infection. One way to improve adherence and decrease the likelihood of drug-drug interactions in HIV-1-infected patients is through the development of long-acting biologic inhibitors. This study describes the development and properties of an adnectin molecule that targets the most conserved region of the gp41 protein and inhibits HIV-1 with good potency. Moreover, when fused to a similar adnectin targeted to the human CD4 protein, the receptor for HIV-1, significant synergies in potency and efficacy are observed. These inhibitors are part of an effort to develop a larger biologic molecule that functions as a long-acting self-administered regimen for patients with HIV-1 infection.


Assuntos
Fármacos Anti-HIV/farmacologia , Antígenos CD4/metabolismo , Proteína gp41 do Envelope de HIV/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Sequência de Aminoácidos , Fármacos Anti-HIV/química , Sítios de Ligação , Linhagem Celular , Técnicas de Visualização da Superfície Celular , Fibronectinas/química , Células HEK293 , Proteína gp41 do Envelope de HIV/química , HIV-1/efeitos dos fármacos , HIV-1/fisiologia , Humanos , Fusão de Membrana/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/química , Produtos do Gene env do Vírus da Imunodeficiência Humana/antagonistas & inibidores
3.
Mol Pharmacol ; 92(3): 310-317, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28645932

RESUMO

The NaV1.7 voltage-gated sodium channel is implicated in human pain perception by genetics. Rare gain of function mutations in NaV1.7 lead to spontaneous pain in humans whereas loss of function mutations results in congenital insensitivity to pain. Hence, agents that specifically modulate the function of NaV1.7 have the potential to yield novel therapeutics to treat pain. The complexity of the channel and the challenges to generate recombinant cell lines with high NaV1.7 expression have led to a surrogate target strategy approach employing chimeras with the bacterial channel NaVAb. In this report we describe the design, synthesis, purification, and characterization of a chimera containing part of the voltage sensor domain 2 (VSD2) of NaV1.7. Importantly, this chimera, DII S1-S4, forms functional sodium channels and is potently inhibited by the NaV1.7 VSD2 targeted peptide toxin ProTx-II. Further, we show by [125I]ProTx-II binding and surface plasmon resonance that the purified DII S1-S4 protein retains high affinity ProTx-II binding in detergent. We employed the purified DII S1-S4 protein to create a scintillation proximity assay suitable for high-throughput screening. The creation of a NaV1.7-NaVAb chimera with the VSD2 toxin binding site provides an important tool for the identification of novel NaV1.7 inhibitors and for structural studies to understand the toxin-channel interaction.


Assuntos
Proteínas de Bactérias/química , Canal de Sódio Disparado por Voltagem NAV1.7/fisiologia , Proteínas Recombinantes de Fusão/química , Venenos de Aranha/metabolismo , Canais de Sódio Disparados por Voltagem/química , Proteínas de Bactérias/fisiologia , Sítios de Ligação , Células HEK293 , Humanos , Ressonância de Plasmônio de Superfície , Canais de Sódio Disparados por Voltagem/fisiologia
4.
Artigo em Inglês | MEDLINE | ID: mdl-28584151

RESUMO

A novel fibronectin-based protein (Adnectin) HIV-1 inhibitor was generated using in vitro selection. This inhibitor binds to human CD4 with a high affinity (3.9 nM) and inhibits viral entry at a step after CD4 engagement and preceding membrane fusion. The progenitor sequence of this novel inhibitor was selected from a library of trillions of Adnectin variants using mRNA display and then further optimized for improved antiviral and physical properties. The final optimized inhibitor exhibited full potency against a panel of 124 envelope (gp160) proteins spanning 11 subtypes, indicating broad-spectrum activity. Resistance profiling studies showed that this inhibitor required 30 passages (151 days) in culture to acquire sufficient resistance to result in viral titer breakthrough. Resistance mapped to the loss of multiple potential N-linked glycosylation sites in gp120, suggesting that inhibition is due to steric hindrance of CD4-binding-induced conformational changes.


Assuntos
Fármacos Anti-HIV/metabolismo , Antígenos CD4/metabolismo , Fibronectinas/metabolismo , Proteína gp120 do Envelope de HIV/metabolismo , Proteína gp160 do Envelope de HIV/antagonistas & inibidores , HIV-1/efeitos dos fármacos , Linhagem Celular , Técnicas de Visualização da Superfície Celular , Epitopos/metabolismo , Glicosilação , Células HEK293 , Humanos , Ligação Proteica
5.
Antiviral Res ; : 105953, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38960100

RESUMO

Temsavir binds directly to the HIV-1 envelope glycoprotein gp120 and selectively inhibits interactions between HIV-1 and CD4 receptors. Previous studies identified gp120 amino acid positions where substitutions are associated with reduced susceptibility to temsavir. The mechanism by which temsavir susceptibility is altered in these envelope glycoproteins was evaluated. Pseudoviruses encoding gp120 substitutions alone (S375H/I/M/N, M426L, M434I, M475I) or in combination (S375H + M475I) were engineered on a wild-type JRFL background. Temsavir-gp120 and CD4-gp120 binding kinetics and ability of temsavir to block CD4-gp120 binding were evaluated using the purified polymorphic gp120 proteins and a Creoptix® WAVE Delta grating-coupled interferometry system. The fold-change in half-maximal inhibitory concentration (IC50) in JRFL-based pseudoviruses containing the aforementioned polymorphisms relative to that of wild-type ranged from 4-fold to 29,726-fold, while temsavir binding affinity for the polymorphic gp120 proteins varied from 0.7-fold to 73.7-fold relative to wild-type gp120. Strong correlations between temsavir IC50 and temsavir binding affinity (r=0.7332; P=0.0246) as well as temsavir binding on-rate (r=-0.8940; P=0.0011) were observed. Binding affinity of gp120 proteins for CD4 varied between 0.4-fold and 3.1-fold compared with wild-type gp120; no correlations between temsavir IC50 and CD4 binding kinetic parameters were observed. For all polymorphic gp120 proteins, temsavir was able to fully block CD4 binding; 3 polymorphs required higher temsavir concentrations. Loss of susceptibility to temsavir observed for gp120 polymorphisms strongly correlated with reductions in temsavir binding on-rate. Nonetheless, temsavir retained the ability to fully block CD4-gp120 engagement given sufficiently high concentrations.

6.
Antivir Ther ; 27(5): 13596535221131164, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36191080

RESUMO

BACKGROUND: The GSK3732394 multivalent protein was developed as a novel, long-acting, antiretroviral biologic treatment regimen with three independent, non-cross-resistant mechanisms for inhibiting HIV-1 entry. METHODS: A single-centre, Phase 1, double-blind, randomized, placebo-controlled study was conducted in healthy volunteers, using a 2-part adaptive study design: in Part 1, participants were randomized to receive subcutaneous injection of GSK3732394 or placebo (3:1) as single ascending doses (10-mg starting dose); in Part 2, participants were intended to receive multiple ascending doses. Primary and secondary objectives included safety, pharmacokinetics (PK) and pharmacodynamics (PD; cluster of differentiation four receptor occupancy [CD4 RO]) of GSK3732394 in healthy adults; PK/PD results in healthy volunteers were used to project HIV-1 treatment success. RESULTS: The most frequently reported adverse event was injection site reactions (ISRs; 8/18 [44%]). Most ISRs were mild (Grade 1-2; n = 7); one participant experienced a Grade 3 ISR (erythema ≥10 cm). All ISRs were delayed in onset (after Day 10). GSK3732394 demonstrated linear PK across all cohorts. Clearance was faster than expected, and PK/PD results were lower than expected, with the maximum dose investigated (80 mg) achieving mean trough CD4 RO of ∼25% on Day 7. The study was terminated as the PK/PD model linking PK and CD4 RO indicated that the maximum planned doses would not achieve the desired therapeutic profile. CONCLUSIONS: This study demonstrated successful deployment of PK/PD dose relationships in the design and conduct of clinical trials by leveraging the findings toward predicting probability of success, resulting in appropriate early termination (ClinicalTrials.gov, NCT03984812).


Assuntos
Produtos Biológicos , Infecções por HIV , HIV-1 , Adulto , Relação Dose-Resposta a Droga , Método Duplo-Cego , Infecções por HIV/tratamento farmacológico , Voluntários Saudáveis , Humanos
7.
J Mol Biol ; 434(2): 167395, 2022 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-34896364

RESUMO

GSK3732394 is a multi-specific biologic inhibitor of HIV entry currently under clinical evaluation. A key component of this molecule is an adnectin (6940_B01) that binds to CD4 and inhibits downstream actions of gp160. Studies were performed to determine the binding site of the adnectin on CD4 and to understand the mechanism of inhibition. Using hydrogen-deuterium exchange with mass spectrometry (HDX), CD4 peptides showed differential rates of deuteration (either enhanced or slowed) in the presence of the adnectin that mapped predominantly to the interface of domains 2 and 3 (D2-D3). In addition, an X-ray crystal structure of an ibalizumab Fab/CD4(D1-D4)/adnectin complex revealed an extensive interface between the adnectin and residues on CD4 domains D2-D4 that stabilize a novel T-shaped CD4 conformation. A cryo-EM map of the gp140/CD4/GSK3732394 complex clearly shows the bent conformation for CD4 while bound to gp140. Mutagenic analyses on CD4 confirmed that amino acid F202 forms a key interaction with the adnectin. In addition, amino acid L151 was shown to be a critical indirect determinant of the specificity for binding to the human CD4 protein over related primate CD4 molecules, as it appears to modulate CD4's flexibility to adopt the adnectin-bound conformation. The significant conformational change of CD4 upon adnectin binding brings the D1 domain of CD4 in proximity to the host cell membrane surface, thereby re-orienting the gp120 binding site in a direction that is inaccessible to incoming virus due to a steric clash between gp160 trimers on the virus surface and the target cell membrane.


Assuntos
Fármacos Anti-HIV/farmacologia , Antígenos CD4/química , Antígenos CD4/metabolismo , HIV-1/metabolismo , Ligação Viral/efeitos dos fármacos , Animais , Anticorpos Monoclonais , Sítios de Ligação , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Domínios Proteicos , Internalização do Vírus/efeitos dos fármacos
8.
Rheumatology (Oxford) ; 50(6): 1033-44, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21258049

RESUMO

OBJECTIVES: To characterize the in vitro binding and effector function properties of CD20-directed small modular immunopharmaceutical (SMIP) 2LM20-4, and to compare its in vivo B-cell depletion activity with the mutated 2LM20-4 P331S [no in vitro complement-dependent cytotoxicity (CDC)] and rituximab in cynomolgus monkeys. METHODS: Direct binding is examined in flow cytometry, confocal microscopy, scatchard and lipid raft assays. Effector function assays include CDC and Fc-mediated cellular toxicity. In the 6-month-long in vivo B-cell depletion study, single i.v. dosages of 1 or 10 mg/kg of anti-CD20 proteins were administered to monkeys and B-cell counts were monitored in peripheral blood, bone marrow and lymph nodes. RESULTS: 2LM20-4 has lower saturation binding to human primary B cells and recruits fewer CD20 molecules into lipid rafts compared with rituximab; however, it induces higher in vitro CDC. In competitive binding, 2LM20-4 only partially displaces rituximab, suggesting that it binds to a fraction of CD20 molecules within certain locations of the plasma membrane as compared with rituximab. In monkeys, 2LM20-4 had more sustained B-cell depletion activity than rituximab in peripheral blood and had significantly more profound and sustained activity than 2LM20-4 P331S and rituximab in the lymph nodes. CONCLUSIONS: SMIP 2LM20-4, which binds to a fraction of CD20 molecules as compared with rituximab, has more potent in vitro CDC, and more potent and sustained B-cell depletion activity in cynomolgus monkeys. Our work has considerable clinical relevance since it provides novel insights related to the emerging B-cell depletion therapies in autoimmune diseases.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Antígenos CD20/efeitos dos fármacos , Antígenos CD20/imunologia , Anticorpos de Cadeia Única/farmacologia , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Murinos/farmacologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Sítios de Ligação , Células Cultivadas , Modelos Animais de Doenças , Humanos , Fatores Imunológicos/farmacologia , Técnicas In Vitro , Modelos Lineares , Macaca fascicularis , Distribuição Aleatória , Rituximab , Sensibilidade e Especificidade
9.
Pharm Res ; 28(7): 1696-706, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21424161

RESUMO

PURPOSE: Anti-Aß Ab2 (Ab2) is a humanized monoclonal antibody against amino acids 3-6 of primate (but not rodent) amyloid ß (Aß) and is being evaluated for the treatment of Alzheimer's disease (AD). This study was conducted to predict the human pharmacokinetics of Ab2. METHODS: In vivo PK profile of Ab2 in preclinical species and in vitro mechanistic studies in preclinical and human systems were used for pharmacokinetic predictions. RESULTS: In Tg2576 and PSAPP mice that have ~100-fold higher circulating levels of human Aß compared to humans, elimination of Ab2 was target-mediated, such that exposure was 5-10 fold lower compared to wild-type rodents or to PDAPP mice that have human Aß concentrations in plasma similar to humans. In cynomolgus monkeys, the t(1/2) of Ab2 was faster (<2.5 days) compared to that of the control antibody (~13 days). The fast elimination of Ab2 in cynomolgus monkeys was linked to off-target binding to cynomolgus monkey fibrinogen that was also causing incomplete recovery of Ab2 in cynomolgus monkey serum in blood partitioning experiments. Ab2 had significantly weaker to undetectable binding to human (and mouse) fibrinogen and had good recovery in human serum in blood partitioning experiments. CONCLUSIONS: These data predict that elimination of Ab2 in healthy or AD humans is expected to be slow, with t(1/2) similar to that observed for other humanized antibodies.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Anticorpos Monoclonais/farmacocinética , Peptídeos beta-Amiloides/sangue , Peptídeos beta-Amiloides/farmacocinética , Peptídeos beta-Amiloides/urina , Animais , Anticorpos Monoclonais/sangue , Anticorpos Monoclonais/urina , Western Blotting , Células CHO , Cricetinae , Cricetulus , Eletroforese em Gel de Poliacrilamida , Feminino , Humanos , Macaca fascicularis , Masculino , Espectrometria de Massas , Camundongos , Camundongos Transgênicos , Fragmentos de Peptídeos/sangue , Fragmentos de Peptídeos/urina , Ratos , Ratos Sprague-Dawley , Ratos Wistar
10.
Biotechnol Bioeng ; 100(5): 839-54, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18551522

RESUMO

High-throughput screening (HTS) of chromatography resins for identifying optimal protein purification conditions is becoming an integral part of industrial process development. In this work, ceramic hydroxyapatite (cHA) chromatography of 15 humanized monoclonal antibodies (mAbs) was examined by HTS. MAb binding, as quantified by partition coefficient (K(p)), was measured under 92 combinations of sodium chloride, phosphate, and pH. Binding varied inversely with these variables for all mAbs tested. However, the magnitudes of binding among mAbs under identical conditions varied significantly, showing a >1.5 log range in K(p). Analysis of variance (ANOVA) techniques were used to describe the binding of each mAb as a function of the three screen variables. Linear models relating log K(p) to the pH, log[sodium chloride], and log[phosphate] fit the data for each antibody with 93-96% accuracy. From these models, characteristic charge values for the cation exchange and metal coordination components of the multi-modal mAb/cHA interaction varied twofold across the mAbs, reflecting inherent variability in the number of contacts between a particular mAb and the cHA surface. Furthermore, we reduced the number of test conditions required from 92 to 8 while maintaining an accurate representation of the full binding response surface. This eight-point modeling method accurately predicted the binding behavior of mAbs as well as mAb aggregates, a common impurity in crude mAb preparations. Using this eight-point modeling method, binding and selectivity information for mAb and aggregate can be obtained from less than two milligrams of protein, making the method attractive for early manufacturability assessments.


Assuntos
Anticorpos Monoclonais/química , Anticorpos Monoclonais/isolamento & purificação , Cerâmica/química , Cromatografia/métodos , Durapatita/química , Microquímica/métodos , Modelos Químicos , Algoritmos , Cromatografia/instrumentação , Simulação por Computador , Sensibilidade e Especificidade
11.
J Mol Biol ; 427(4): 924-942, 2015 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-25579995

RESUMO

The human pregnane X receptor (PXR) is a promiscuous nuclear receptor that functions as a sensor to a wide variety of xenobiotics and regulates expression of several drug metabolizing enzymes and transporters. We have generated "Adnectins", derived from 10th fibronectin type III domain ((10)Fn3), that target the PXR ligand binding domain (LBD) interactions with the steroid receptor co-activator-1 (SRC-1) peptide, displacing SRC-1 binding. Adnectins are structurally homologous to the immunoglobulin superfamily. Three different co-crystal structures of PXR LBD with Adnectin-1 and CCR1 (CC chemokine receptor-1) antagonist Compound-1 were determined. This structural information was used to modulate PXR affinity for a related CCR1 antagonist compound that entered into clinical trials for rheumatoid arthritis. The structures of PXR with Adnectin-1 reveal specificity of Adnectin-1 in not only targeting the interface of the SRC-1 interactions but also engaging the same set of residues that are involved in binding of SRC-1 to PXR. Substituting SRC-1 with Adnectin-1 does not alter the binding conformation of Compound-1 in the ligand binding pocket. The structure also reveals the possibility of using Adnectins as crystallization chaperones to generate structures of PXR with compounds of interest.


Assuntos
Coativador 1 de Receptor Nuclear/química , Receptores CCR1/antagonistas & inibidores , Receptores de Esteroides/química , Ureia/análogos & derivados , Valina/análogos & derivados , Sequência de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Humanos , Lignanas/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Receptor de Pregnano X , Ligação Proteica , Estrutura Terciária de Proteína , Receptores CCR1/metabolismo , Alinhamento de Sequência , Ressonância de Plasmônio de Superfície , Ureia/química , Ureia/metabolismo , Ureia/farmacologia , Valina/química , Valina/metabolismo , Valina/farmacologia
12.
Expert Opin Drug Saf ; 7(1): 5-8, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18171310

RESUMO

Methadone is an interesting analgesic for multiple reasons. The unique properties of the agent, low cost and widespread availability have led to increases in methadone prescribing. Despite advantages, methadone is challenging to work with, particularly in patients with high opioid requirements. Recent concerns regarding cardiac arrhythmias and respiratory depression have led to changes in the labeling of methadone. This editorial highlights some of these concerns and provides some recommendations for the appropriate use of methadone in the setting of pain.


Assuntos
Metadona/efeitos adversos , Animais , Humanos , Metadona/uso terapêutico , Transtornos Relacionados ao Uso de Opioides/metabolismo , Transtornos Relacionados ao Uso de Opioides/prevenção & controle , Dor/tratamento farmacológico , Dor/metabolismo , Insuficiência Respiratória/induzido quimicamente , Insuficiência Respiratória/prevenção & controle
13.
J Virol ; 77(6): 3460-9, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12610121

RESUMO

During ongoing C-type retrovirus infection, the probability of leukemia caused by insertional gene activation is markedly increased by the emergence of recombinant retroviruses that repeatedly infect host cells. The murine mink cell focus-inducing (MCF) viruses with this property have acquired characteristic changes in the N-terminal domain of their envelope glycoprotein that specify binding to a different receptor than the parental ecotropic virus. In this report, we show that MCF virus infection occurs through binding to this receptor (termed Syg1) and, remarkably, by a second mechanism that does not utilize the Syg1 receptor. By the latter route, the N-terminal domain of the ecotropic virus glycoprotein expressed on the cell surface in a complex with its receptor activates the fusion mechanism of the MCF virus in trans. The rate of MCF virus spread through a population of permissive human cells was increased by establishment of trans activation, indicating that Syg1 receptor-dependent and -independent pathways function in parallel. Also, trans activation shortened the interval between initial infection and onset of cell-cell fusion associated with repeated infection of the same cell. Our findings indicate that pathogenic retrovirus infection may be initiated by virus binding to cell receptors or to the virus envelope glycoprotein of other viruses expressed on the cell surface. Also, they support a broader principle: that cooperative virus-virus interactions, as well as virus-host interactions, shape the composition and properties of the retrovirus quasispecies.


Assuntos
Fusão de Membrana , Vírus Indutores de Focos em Células do Vison/metabolismo , Vírus Indutores de Focos em Células do Vison/patogenicidade , Receptores Virais/metabolismo , Transdução de Sinais , Proteínas do Envelope Viral/metabolismo , Animais , Fusão Celular , Linhagem Celular , Cricetinae , Regulação Viral da Expressão Gênica , Humanos , Camundongos , Ativação Transcricional , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Receptor do Retrovírus Politrópico e Xenotrópico
14.
J Virol ; 77(4): 2717-29, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12552012

RESUMO

Infection of T lymphocytes by the cytopathic retrovirus feline leukemia virus subgroup T (FeLV-T) requires FeLIX, a cellular coreceptor that is encoded by an endogenous provirus and closely resembles the receptor-binding domain (RBD) of feline leukemia virus subgroup B (FeLV-B). We determined the structure of FeLV-B RBD, which has FeLIX activity, to a 2.5-A resolution by X-ray crystallography. The structure of the receptor-specific subdomain of this glycoprotein differs dramatically from that of Friend murine leukemia virus (Fr-MLV), which binds a different cell surface receptor. Remarkably, we find that Fr-MLV RBD also activates FeLV-T infection of cells expressing the Fr-MLV receptor and that FeLV-B RBD is a competitive inhibitor of infection under these conditions. These studies suggest that FeLV-T infection relies on the following property of mammalian leukemia virus RBDs: the ability to couple interaction with one of a variety of receptors to the activation of a conserved membrane fusion mechanism. A comparison of the FeLV-B and Fr-MLV RBD structures illustrates how receptor-specific regions are linked to conserved elements critical for postbinding events in virus entry.


Assuntos
Vírus da Leucemia Felina/patogenicidade , Receptores Virais/química , Receptores Virais/metabolismo , Sequência de Aminoácidos , Animais , Gatos , Linhagem Celular , Cricetinae , Cristalização , Cristalografia por Raios X , Glicoproteínas/química , Humanos , Vírus da Leucemia Felina/metabolismo , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Alinhamento de Sequência , Proteínas do Envelope Viral/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA