Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
2.
Sci Rep ; 9(1): 17830, 2019 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-31780710

RESUMEN

Programmed cell death 1 (PD-1) monoclonal antibodies have been approved by regulatory agencies for the treatment of various types of cancer, and the mechanism involves the restoration of T cell functions. We report herein the X-ray crystal structure of a fully human monoclonal antibody mAb059c fragment antigen-binding (Fab) in complex with the PD-1 extracellular domain (ECD) at a resolution of 1.70 Å. Structural analysis indicates 1) an epitope, comprising fragments from the C'D, BC and FG loops of PD-1, contributes to mAb059c interaction, 2) an unique conformation of the C'D loop and a different orientation of R86 enabling the capture of PD-1 by the antibody complementarity determining region (CDR) and the formation of one salt-bridge contact - ASP101(HCDR3):ARG86(PD-1), and 3) the contact of FG with light chain (LC) CDR3 is maintained by a second salt-bridge and two backbone hydrogen bonds. Interface analysis reveals that N-glycosylation sites 49, 74 and 116 on PD-1 do not contact mAb059c; while N58 in the BC loop is recognized by mAb059c heavy chain CDR1 and CDR2. Mutation of N58 attenuated mAb059c binding to PD-1. These findings and the novel anti-PD-1 antibody will facilitate better understanding of the mechanisms of the molecular recognition of PD-1 receptor by anti-PD-1 mAb and, thereby, enable the development of new therapeutics with an expanded spectrum of efficacy for unmet medical needs.


Asunto(s)
Adenocarcinoma/terapia , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/uso terapéutico , Neoplasias del Colon/terapia , Receptor de Muerte Celular Programada 1/química , Animales , Afinidad de Anticuerpos , Complejo Antígeno-Anticuerpo/química , Línea Celular Tumoral , Modelos Animales de Enfermedad , Epítopos/química , Técnicas de Sustitución del Gen , Células HEK293 , Humanos , Enlace de Hidrógeno , Fragmentos Fab de Inmunoglobulinas/química , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptor de Muerte Celular Programada 1/genética , Unión Proteica , Dominios Proteicos , Transfección
3.
Microbes Infect ; 20(9-10): 626-634, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29126877

RESUMEN

Although combination antiretroviral therapy (cART) is highly effective in suppressing human immunodeficiency virus type 1 (HIV-1) replication, it fails to eradicate the virus from HIV-1-infected individuals because HIV-1 integrates into the resting CD4+ T cells, establishing latently infected reservoirs. Histone deacetylation is a key element in regulating HIV-1 latent infection. Chidamide, a new anticancer drug, is a novel type of selective histone deacetylase inhibitor. Here we showed that chidamide effectively reactivated HIV-1 latent provirus in different latently infected cell lines in a dose- and time-dependent manner. Chidamide had relatively low cytotoxicity to peripheral blood mononuclear cells (PBMCs) and other latent cell lines. We have demonstrated that chidamide reactivated HIV-1 latent provirus through the NF-κB signaling pathway. The replication of the newly reactivated HIV-1 could then be effectively inhibited by the anti-HIV-1 drugs Zidovudine, Nevirapine, and Indinavir. Therefore, chidamide might be used in combination with cART for functional HIV-1 cure.


Asunto(s)
Aminopiridinas/farmacología , Benzamidas/farmacología , VIH-1/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Provirus/efectos de los fármacos , Activación Viral/efectos de los fármacos , Latencia del Virus/efectos de los fármacos , Fármacos Anti-VIH/farmacología , Línea Celular , Supervivencia Celular , Infecciones por VIH/virología , VIH-1/fisiología , Humanos , Cinética , FN-kappa B/metabolismo , Provirus/fisiología , Transducción de Señal , Replicación Viral/efectos de los fármacos
4.
Biosci Trends ; 11(4): 450-459, 2017 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-28652534

RESUMEN

Respiratory Syncycial Virus (RSV) is the most important pathogen responsible for children's severe lower respiratory tract infection. So far no RSV vaccine has yet been authorized for clinical use. The main impediment that blocked development of RSV vaccine is that inactivated RSV vaccine could cause RSV vaccine-enhanced disease (RVED). The mechanism of RVED remains unclear. Recently some researchers found that insufficient activation of innate immunity, including Toll-like receptors (TLRs), might be associated with the onset of RVED. Based on the above findings, this research was conducted to further study the mechanism of RVED. We first vaccinated mice with formalin-inactivated RSV vaccine (FIRSV) and then exposed them to RSV to establish a RVED mouse model. Consequently, we found that mice previously inoculated with FIRSV showed obvious weight loss and extensive pneumonia, as well as T helper 2 cells (Th2)-biased immunity and suppressed CD8+T cell immunity after viral exposure, suggesting that we have successfully established a RVED mouse model. Then based on this model, we further added Poly(U) (TLR7/8 agonist) and CpG (TLR9 agonist) in FIRSV to see if RVED could be ameliorated. As a result, mice inoculated with FIRSV supplemented with Poly(U) and CpG had a much relieved weight loss and pneumonia, as well as suppressed Th2-biased immunity and strengthened CD8+T cell function. Thus, the insufficient stimulation of TLR7/8 and (or) TLR9 might play a role in the development of RVED, which could provide evidence for using TLR agonists as vaccine adjuvants to confer a protective immune response against RSV.


Asunto(s)
Inmunidad/efectos de los fármacos , Oligodesoxirribonucleótidos/uso terapéutico , Neumonía/inmunología , Neumonía/virología , Vacunas contra Virus Sincitial Respiratorio/efectos adversos , Linfocitos T/inmunología , Uracilo/uso terapéutico , Animales , Peso Corporal/efectos de los fármacos , Femenino , Formaldehído , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Ratones Endogámicos BALB C , Oligodesoxirribonucleótidos/farmacología , Neumonía/patología , Virus Sincitiales Respiratorios/efectos de los fármacos , Virus Sincitiales Respiratorios/fisiología , Linfocitos T/efectos de los fármacos , Receptores Toll-Like/agonistas , Receptores Toll-Like/metabolismo , Uracilo/farmacología , Vacunas de Productos Inactivados/inmunología , Carga Viral/efectos de los fármacos
5.
Cell Host Microbe ; 22(4): 471-483.e5, 2017 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-28966056

RESUMEN

The H7N9 influenza virus causes high-mortality disease in humans but no effective therapeutics are available. Here we report a human monoclonal antibody, m826, that binds to H7 hemagglutinin (HA) and protects against H7N9 infection. m826 binds to H7N9 HA with subnanomolar affinity at acidic pH and 10-fold lower affinity at neutral pH. The high-resolution (1.9 Å) crystal structure of m826 complexed with H7N9 HA indicates that m826 binds an epitope that may be fully exposed upon pH-induced conformational changes in HA. m826 fully protects mice against lethal challenge with H7N9 virus through mechanisms likely involving antibody-dependent cell-mediated cytotoxicity. Interestingly, immunogenetic analysis indicates that m826 is a germline antibody, and m826-like sequences can be identified in H7N9-infected patients, healthy adults, and newborn babies. These m826 properties offer a template for H7N9 vaccine immunogens, a promising candidate therapeutic, and a tool for exploring mechanisms of virus infection inhibition by antibodies.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Epítopos/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H7N9 del Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/química , Perros , Femenino , Células HEK293 , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Subtipo H7N9 del Virus de la Influenza A/química , Vacunas contra la Influenza/inmunología , Gripe Humana/terapia , Gripe Humana/virología , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos BALB C , Conformación Molecular , Infecciones por Orthomyxoviridae/terapia , Infecciones por Orthomyxoviridae/virología
6.
Emerg Microbes Infect ; 5: e65, 2016 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-27329850

RESUMEN

After three decades of intensive research efforts, an effective vaccine against HIV-1 remains to be developed. Several broadly neutralizing antibodies to HIV-1, such as 10E8, recognize the membrane proximal external region (MPER) of the HIV-1 gp41 protein. Thus, the MPER is considered to be a very important target for vaccine design. However, the MPER segment has very weak immunogenicity and tends to insert its epitope residues into the cell membrane, thereby avoiding antibody binding. To address this complication in vaccine development, we herein designed a peptide, designated 10E8-4P, containing four copies of the 10E8 epitope as an immunogen. As predicted by structural simulation, 10E8-4P exhibits a well-arranged tandem helical conformation, with the key residues in the 10E8 epitope oriented at different angles, thus suggesting that some of these key residues may be exposed outside of the lipid membrane. Compared with a peptide containing a single 10E8 epitope (10E8-1P), 10E8-4P not only exhibited better antigenicity but also elicited neutralizing antibody response against HIV-1 pseudoviruses, whereas 10E8-1P could not induce detectable neutralizing antibody response. Importantly, antibodies elicited by 10E8-4P also possessed a strong ability to activate an antibody-dependent cell-mediated cytotoxicity (ADCC) reporter gene, thus suggesting that they may have ADCC activity. Therefore, this strategy shows promise for further optimization and application in future HIV-1 vaccine design.


Asunto(s)
Vacunas contra el SIDA/inmunología , Anticuerpos Neutralizantes/biosíntesis , Citotoxicidad Celular Dependiente de Anticuerpos/genética , Epítopos/inmunología , Anticuerpos Anti-VIH/inmunología , VIH-1/inmunología , Vacunas contra el SIDA/química , Animales , Dicroismo Circular , Ensayo de Inmunoadsorción Enzimática , Epítopos/química , Genes Reporteros , Anticuerpos Anti-VIH/biosíntesis , Humanos , Pruebas de Neutralización , Unión Proteica , Conejos
7.
Viruses ; 7(2): 798-819, 2015 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-25690799

RESUMEN

Respiratory syncytial virus (RSV) is the leading cause of pediatric viral respiratory tract infections. Neither vaccine nor effective antiviral therapy is available to prevent and treat RSV infection. Palivizumab, a humanized monoclonal antibody, is the only product approved to prevent serious RSV infection, but its high cost is prohibitive in low-income countries. Here, we aimed to identify an effective, safe, and affordable antiviral agent for pre-exposure prophylaxis (PrEP) of RSV infection in children at high risk. We found that maleic anhydride (ML)-modified human serum albumin (HSA), designated ML-HSA, exhibited potent antiviral activity against RSV and that the percentages of the modified lysines and arginies in ML- are correlated with such anti-RSV activity. ML-HSA inhibited RSV entry and replication by interacting with viral G protein and blocking RSV attachment to the target cells, while ML-HAS neither bound to F protein, nor inhibited F protein-mediated membrane fusion. Intranasal administration of ML-HSA before RSV infection resulted in significant decrease of the viral titers in the lungs of mice. ML-HSA shows promise for further development into an effective, safe, affordable, and easy-to-use intranasal regimen for pre-exposure prophylaxis of RSV infection in children at high risk in both low- and high-income countries.


Asunto(s)
Antivirales/administración & dosificación , Anhídridos Maleicos , Profilaxis Pre-Exposición , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Albúmina Sérica/administración & dosificación , Administración Intranasal , Animales , Antivirales/química , Antivirales/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Anhídridos Maleicos/química , Ratones , Pruebas de Sensibilidad Microbiana , Unión Proteica , Albúmina Sérica/química , Albúmina Sérica/metabolismo , Proteínas del Envoltorio Viral/metabolismo
8.
Sci Rep ; 5: 13028, 2015 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-26286358

RESUMEN

Enfuvirtide (T20), is the first HIV fusion inhibitor approved for treatment of HIV/AIDS patients who fail to respond to the current antiretroviral drugs. However, its clinical application is limited because of short half-life, drug resistance and cross-reactivity with the preexisting antibodies in HIV-infected patients. Using an artificial peptide strategy, we designed a peptide with non-native protein sequence, AP3, which exhibited potent antiviral activity against a broad spectrum of HIV-1 strains, including those resistant to T20, and had remarkably longer in vivo half-life than T20. While the preexisting antibodies in HIV-infected patients significantly suppressed T20's antiviral activity, these antibodies neither recognized AP3, nor attenuated its anti-HIV-1 activity. Structurally different from T20, AP3 could fold into single-helix and interact with gp41 NHR. The two residues, Met and Thr, at the N-terminus of AP3 form a hook-like structure to stabilize interaction between AP3 and NHR helices. Therefore, AP3 has potential for further development as a new HIV fusion inhibitor with improved antiviral efficacy, resistance profile and pharmacological properties over enfuvirtide. Meanwhile, this study highlighted the advantages of artificially designed peptides, and confirmed that this strategy could be used in developing artificial peptide-based viral fusion inhibitors against HIV and other enveloped viruses.


Asunto(s)
Proteína gp41 de Envoltorio del VIH/farmacología , Inhibidores de Fusión de VIH/química , Inhibidores de Fusión de VIH/farmacología , Fragmentos de Péptidos/farmacología , Péptidos/farmacología , Secuencia de Aminoácidos , Animales , Fenómenos Biofísicos/efectos de los fármacos , Cristalografía por Rayos X , Endopeptidasa K/metabolismo , Enfuvirtida , Anticuerpos Anti-VIH/inmunología , Proteína gp41 de Envoltorio del VIH/metabolismo , Inhibidores de Fusión de VIH/farmacocinética , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , VIH-1/inmunología , VIH-1/aislamiento & purificación , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/química , Estructura Secundaria de Proteína , Proteolisis/efectos de los fármacos , Ratas Sprague-Dawley
9.
AIDS Res Hum Retroviruses ; 30(2): 160-4, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23800338

RESUMEN

To elucidate the prevalence of HIV-1 subtypes and transmitted drug resistance in Henan, central China, HIV-1-positive blood samples from 187 antiretroviral-naive patients were collected in our study from August 2009 to November 2010. Subtype B' (92.0%, 172 of 187) remains the predominant HIV-1 subtype in Henan province and was prevalent in all risk populations and geographic regions. Of 98 pol sequences 67 (68.4%) harbored drug resistance mutations, and only 14 (14.3%, 14 of 98) sequences have mutations associated with significantly reduced phenotypic susceptibility to antiretroviral drugs. The unexpectedly high percentage of drug resistance in Henan province is mainly due to the prevalence of minor mutations in the protease and integrase regions, especially A71T/V and L68V/I/IM/LV. In all, we detected a relatively high prevalence of drug resistance with unique mutation distributions among antiretroviral-naive patients from Henan province.


Asunto(s)
Farmacorresistencia Viral , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , VIH-1/aislamiento & purificación , Adolescente , Adulto , Anciano , Niño , Preescolar , China , Análisis por Conglomerados , Femenino , Genotipo , VIH-1/clasificación , VIH-1/genética , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Mutación Missense , Prevalencia , Análisis de Secuencia de ADN , Adulto Joven , Productos del Gen pol del Virus de la Inmunodeficiencia Humana/genética
10.
Virus Res ; 194: 200-10, 2014 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-25451066

RESUMEN

The recent outbreak of Middle East respiratory syndrome (MERS) coronavirus (MERS-CoV) infection has led to more than 800 laboratory-confirmed MERS cases with a high case fatality rate (∼35%), posing a serious threat to global public health and calling for the development of effective and safe therapeutic and prophylactic strategies to treat and prevent MERS-CoV infection. Here we discuss the most recent studies on the structure of the MERS-CoV spike protein and its role in virus binding and entry, and the development of MERS-CoV entry/fusion inhibitors targeting the S1 subunit, particularly the receptor-binding domain (RBD), and the S2 subunit, especially the HR1 region, of the MERS-CoV spike protein. We then look ahead to future applications of these viral entry/fusion inhibitors, either alone or in combination with specific and nonspecific MERS-CoV replication inhibitors, for the treatment and prevention of MERS-CoV infection.


Asunto(s)
Antivirales/farmacología , Coronavirus del Síndrome Respiratorio de Oriente Medio/efectos de los fármacos , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Glicoproteína de la Espiga del Coronavirus/antagonistas & inhibidores , Acoplamiento Viral/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Antivirales/uso terapéutico , Infecciones por Coronavirus/tratamiento farmacológico , Humanos
11.
Viruses ; 5(1): 211-25, 2013 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-23325327

RESUMEN

Human respiratory syncytial virus (RSV) is the main viral cause of respiratory tract infection in infants as well as some elderly and high-risk adults with chronic pulmonary disease and the severely immunocompromised. So far, no specific anti-RSV therapeutics or effective anti-RSV vaccines have been reported. Only one humanized monoclonal antibody, Palivizumab, has been approved for use in high-risk infants to prevent RSV infection. Ribavirin is the only drug licensed for therapy of RSV infection, but its clinical use is limited by its nonspecific anti-RSV activity, toxic effect, and relatively high cost. Therefore, development of novel effective anti-RSV therapeutics is urgently needed. The RSV envelope glycoprotein F plays an important role in RSV fusion with, and entry into, the host cell and, consequently, serves as an attractive target for developing RSV entry inhibitors. This article reviews advances made in studies of the structure and function of the F protein and the development of RSV entry inhibitors targeting it.


Asunto(s)
Antivirales/farmacología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Virus Sincitial Respiratorio Humano/fisiología , Proteínas Virales de Fusión/metabolismo , Internalización del Virus/efectos de los fármacos , Animales , Humanos , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Virus Sincitial Respiratorio Humano/genética , Proteínas Virales de Fusión/antagonistas & inhibidores , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/genética
12.
Microbes Infect ; 15(6-7): 432-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23628410

RESUMEN

Very recently, a new avian flu outbreak in humans, which is caused by a novel H7N9 influenza A virus (AIV), was reported in China. As of April 13, 2013, 49 confirmed cases (mainly middle-aged to elderly males), including 11 deaths, were reported in China. Here we analyzed the genomic signatures and protein sequences of the human H7N9 AIVs. We found that the genomic signatures of A(H7N9) had high and low identity to avian and human IAVs, respectively, suggesting its avian origin. The signature amino acids of A(H7N9) had high identity to 1997 H5N1 and 2009 H1N1, but low identity to those influenza strains that caused pandemics before 1980. One of the key signature amino acids at 627 in PB2 mutated to lysine, which is associated with mammalian adaptation and increased virulence of the highly pathogenic avian influenza A(H5N1) virus. Besides, several other human-like signatures, including PB2-44S, PA-100A, PA-356R, and PA-409N are also found in this avian-origin A(H7N9) virus. The HA protein has the Q226L mutation, which is associated with increased binding to mammalian-like receptors bearing alpha 2,6 receptor in the human upper airway. The M2 protein contains the N31S mutation, suggesting its resistance to the M2 channel blockers amantadine and rimantadine. These findings suggest that this avian-origin AIV gains its bird-to-human, i.e., zoonotic, transmissibility and increased virulence, as well as drug-resistance, by mutating key signature amino acid residues and those in the functional domains of the viral proteins. Therefore, it is prudent to monitor the evolution of A(H7N9), as well as develop strategies to combat any potential epidemic or pandemic.


Asunto(s)
Genoma Viral , Subtipo H7N9 del Virus de la Influenza A/genética , Gripe Humana/virología , ARN Viral/genética , Análisis de Secuencia de ADN , China/epidemiología , Brotes de Enfermedades , Humanos , Subtipo H7N9 del Virus de la Influenza A/aislamiento & purificación , Subtipo H7N9 del Virus de la Influenza A/patogenicidad , Mutación Missense , Proteínas Virales/genética , Virulencia
13.
AIDS Res Hum Retroviruses ; 28(10): 1352-6, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22264007

RESUMEN

The distribution of HIV-1 subtypes and genetic characterization of CRF01_AE in Guangxi, southern China were identified. The distribution of HIV-1 genotypes based on gag, pol, and partial env sequences (n=349) was as follows: CRF01_AE (66.5%), CRF08_BC (19.2%), CRF07_BC (7.2%), URF (4.6%), subtype B (1.7%), and subtype B' (0.9%). CRF01_AE predominated in all geographic regions and risk populations and there were multiple introductions of CRF01_AE strains in Guangxi. We found a peculiar CRF01_AE monophyletic lineage distinct from other CRF01_AE viruses, and we designated it "CRF01_AE-v" for convenience. CRF01_AE-v circulating in both heterosexuals and injecting drug users (IDUs) had accounted for 39.7% of CRF01_AE. It showed a selective advantage in the Guangxi population and formed its own characteristic compared with all the CRF01_AE references. Our results suggested that CRF01_AE-v was a new variant of CRF01_AE and it might lead to a new epidemic in Guangxi.


Asunto(s)
Genes env/genética , Genes gag/genética , Genes pol/genética , Infecciones por VIH/epidemiología , Infecciones por VIH/virología , VIH-1/genética , Abuso de Sustancias por Vía Intravenosa/epidemiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , China/epidemiología , Brotes de Enfermedades , Femenino , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Epidemiología Molecular , Datos de Secuencia Molecular , Embarazo , Conducta Sexual , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA