Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Clin Exp Rheumatol ; 40(7): 1393-1402, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34796837

RESUMEN

OBJECTIVES: In early rheumatoid arthritis (eRA) plasma levels of specific chemokines have been shown to correlate with disease activity. However, it is unclear whether pre-treatment chemokine levels can predict disease remission at week 24, and it is not known how biological treatments with different modes of action affect plasma chemokine levels in patients with untreated eRA. METHODS: This study included 347 Swedish patients with untreated eRA from the larger NORD-STAR randomised treatment trial. Here, eRA patients were treated with methotrexate combined with either prednisolone, anti-TNF (certolizumab-pegol), CTLA-4Ig (abatacept) or anti-IL6 receptor (tocilizumab). The primary clinical outcome was remission by clinical disease activity index (CDAI) defined as CDAI ≤ 2.8. Disease activity was assessed by CDAI, DAS28-ESR, DAS28-CRP, swollen joint counts, tender joint counts, ESR and CRP. The plasma concentrations of 14 chemokines were measured at baseline and after 24 weeks of treatment by bead-based immunoassay or ELISA. RESULTS: Baseline plasma concentrations of CXCL10, CXCL8, CXCL9, CXCL11, CXCL5 and CCL2 correlated with baseline disease activity measures. After 24 weeks of treatment, plasma levels of CXCL10, CXCL8, CXCL9, CXCL11 and CXCL13 decreased in all treatment groups except in patients treated with anti-IL6 receptor. In multivariate factor analysis, plasma chemokine levels at baseline could not differentiate patients who attained remission by week 24 from those who did not in any of the treatment groups. CONCLUSIONS: In patients with untreated eRA, plasma levels of several chemokines correlate with disease activity at baseline but cannot predict remission after 24 weeks of treatment with methotrexate combined with prednisolone, anti­TNF, CTLA­4Ig or anti­IL6R.


Asunto(s)
Antirreumáticos , Artritis Reumatoide , Antirreumáticos/uso terapéutico , Artritis Reumatoide/diagnóstico , Artritis Reumatoide/tratamiento farmacológico , Biomarcadores , Humanos , Metotrexato/uso terapéutico , Prednisolona/uso terapéutico , Inducción de Remisión , Índice de Severidad de la Enfermedad , Resultado del Tratamiento , Inhibidores del Factor de Necrosis Tumoral
2.
Immunity ; 30(3): 324-35, 2009 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-19303387

RESUMEN

Follicular helper T (Tfh) cells are the class of effector T helper cells that regulates the step-wise development of antigen-specific B cell immunity in vivo. Deployment of CXCR5+ Tfh cells to B cell zones of lymphoid tissues and stable cognate interactions with B cells are central to the delivery of antigen-specific Tfh cell function. Here, we review recent advances that have helped to unravel distinctive elements of developmental programming for Tfh cells and unique effector Tfh cell functions focused on antigen-primed B cells. Understanding the regulatory functions of Tfh cells in the germinal center and the subsequent regulation of memory B cell responses to antigen recall represent the frontiers of this research area with the potential to alter fundamentally the design of future vaccines.


Asunto(s)
Subgrupos Linfocitarios/citología , Linfocitos T Colaboradores-Inductores/citología , Animales , Linfocitos B/inmunología , Diferenciación Celular , Linaje de la Célula , Humanos , Inmunidad , Subgrupos Linfocitarios/inmunología , Receptores CXCR5/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología
3.
Immunity ; 28(5): 698-709, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18450485

RESUMEN

How T cell receptor (TCR) specificity evolves in vivo after protein vaccination is central to the development of helper T (Th) cell function. Most models of clonal selection in the Th cell compartment favor TCR affinity-based thresholds. Here, we demonstrated that depot-forming vaccine adjuvants did not require Toll-like receptor (TLR) agonists to induce clonal dominance in antigen-specific Th cell responses. However, readily dispersible adjuvants using TLR-9 and TLR-4 agonists skewed TCR repertoire usage by increasing TCR selection thresholds and enhancing antigen-specific clonal expansion. In this manner, vaccine adjuvants control the local accumulation of Th cells expressing TCR with the highest peptide MHC class II binding. Clonal composition was altered by mechanisms that blocked the local propagation of clonotypes independently of antigen dose and not as a consequence of interclonal competition. This capacity of adjuvants to modify antigen-specific Th cell clonal composition has fundamental implications for the design of future protein subunit vaccines.


Asunto(s)
Adyuvantes Inmunológicos , Citocromos c/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Receptores Toll-Like/metabolismo , Vacunas/inmunología , Animales , Citocromos c/inmunología , Activación de Linfocitos , Ratones , Ratones Congénicos , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Receptores Toll-Like/agonistas , Receptores Toll-Like/inmunología , Vacunas de Subunidad/inmunología
4.
Arthritis Res Ther ; 22(1): 245, 2020 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-33066816

RESUMEN

BACKGROUND: The majority of CD4+ T helper (Th) cells found in the synovial fluid (SF) of patients with rheumatoid arthritis (RA) express CXCR3, a receptor associated with Th1 cells. In blood, subsets of Th2 and Th17 cells also express CXCR3, but it is unknown if these cells are present in RA SF or how cytokines from these subsets affect cytokine/chemokine secretion by fibroblast-like synoviocytes (FLS) from patients with RA. METHODS: We examined the proportions of Th1, Th2, CXCR3+Th2, Th17, CXCR3+Th17, Th1Th17, peripheral T helper (TPh) and T follicular helper (TFh) cells in paired SF and blood, as well as the phenotype of TPh and TFh cells in RA SF (n = 8), by the use of flow cytometry. We also examined the cytokine/chemokine profile in paired SF and plasma (n = 8) and in culture supernatants of FLS from patients with chronic RA (n = 7) stimulated with Th-associated cytokines, by the use of cytometric bead arrays and ELISA. Cytokine receptor expression in FLS (n = 3) were assessed by the use of RNA sequencing and qPCR. RESULTS: The proportions of Th1 and CXCR3+Th2 cells were higher in SF than in blood (P < 0.05). TPh and PD-1highTFh in RA SF were primarily of a Th1 and a CXCR3+Th2 phenotype. Moreover, the levels of CXCL9, CXCL10, CCL20, CCL2, CXCL8, IL-6 and IL-10 were higher in SF than in plasma (P < 0.05). Lastly, IL-4, IL-13 and IL-17A induced RA FLS to secrete proinflammatory IL-6, CCL2, CXCL1 and CXCL8, while IFNγ mainly induced CXCL10. CONCLUSION: These findings indicate that not only Th1 but also CXCR3+Th2 cells may have a pathogenic role in RA synovial inflammation.


Asunto(s)
Artritis Reumatoide , Líquido Sinovial , Humanos , Fenotipo , Receptores CXCR3 , Células TH1 , Células Th17 , Células Th2
5.
Immunology ; 123(2): 228-38, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17764451

RESUMEN

Complement is an essential part of the innate immune system, which clears pathogens without requirement for previous exposure, although it also greatly enhances the efficacy and response of the cellular and humoral immune systems. Kaposi's sarcoma-associated herpesvirus (KSHV) is the most recently identified human herpesvirus and the likely aetiological agent of Kaposi's sarcoma, primary effusion lymphoma and multicentric Castleman's disease. We previously reported that the KSHV complement control protein (KCP) was expressed on infected cells and virions, and could inhibit complement through decay-accelerating activity (DAA) of the classical C3 convertase and cofactor activity (CFA) for factor I (FI)-mediated degradation of C4b and C3b, as well as acting as an attachment factor for binding to heparan sulphate on permissive cells. Here, we determined the ability of a panel of monoclonal anti-KCP antibodies to block KCP functions relative to their recognized epitopes, as determined through binding to recombinant KCP containing large (entire domain) or small (2-3 amino acid residue) alterations. One antibody recognizing complement control protein (CCP) domain 1 blocked heparin binding, DAA and C4b CFA, but was poor at blocking C3b CFA, while a second antibody recognizing CCP4 blocked C3b CFA and 80% DAA, but not C4b CFA or heparan sulphate binding. Two antibodies recognizing CCP2 and CCP3 were capable of blocking C3b and C4b CFA and heparan sulphate binding, but only one could inhibit DAA. These results show that, while KCP is a multifunctional protein, these activities do not completely overlap and can be isolated through incubation with monoclonal antibodies.


Asunto(s)
Antígenos CD55/inmunología , Herpesvirus Humano 8/inmunología , Proteínas Virales/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Antitrombina III/inmunología , Unión Competitiva/inmunología , Células CHO , C3 Convertasa de la Vía Clásica del Complemento/inmunología , Complemento C3b/inmunología , Complemento C4b/inmunología , Cricetinae , Cricetulus , Mapeo Epitopo/métodos , Heparitina Sulfato/metabolismo , Ratones , Ratones Endogámicos BALB C
6.
Mol Immunol ; 44(1-3): 11-22, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16905191

RESUMEN

The complement system is an important part of innate immunity providing immediate protection against pathogens without a need for previous exposure, as well as priming the adaptive immune response through opsonisation, leukocyte recruitment and enhancing humoral immune responses. Its importance is not only shown through recurring fulminant infections in individuals with complement component deficiencies, but also through the many complement evasion strategies discovered for a wide range of infectious microbes (including acquisition of endogenous host complement inhibitors and expression of own homologues). Knowledge of these mechanisms at a molecular level may aid development of vaccines and novel therapeutic strategies. Here, we review the structure-function studies of the membrane-bound complement inhibitor KCP that is expressed on the surface of Kaposi's sarcoma-associated herpesvirus (KSHV) virions and infected cells. KCP accelerates the decay of classical C3 convertase and induces the degradation of activated complement factors C4b and C3b by a serine proteinase, factor I. Molecular modeling and site-directed mutagenesis have identified sites on the surface of KCP required for complement inhibition and support the hypothesis that KCP has evolved to mimic the structure and function of endogenous human inhibitors. KCP additionally enhances virion binding to permissive cells through a heparin/heparan sulfate-binding site located at the N-terminus of the protein.


Asunto(s)
Convertasas de Complemento C3-C5/antagonistas & inhibidores , Herpesvirus Humano 8/patogenicidad , Proteínas Virales/fisiología , Aminoácidos/análisis , Heparina/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Inmunidad Innata , Proteínas Virales/análisis , Proteínas Virales/química
7.
Mol Immunol ; 43(10): 1665-75, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16442624

RESUMEN

The Kaposi's sarcoma-associated herpesvirus (KSHV) complement control protein (KCP) inhibits the human complement system, and is similar in structure and function to endogenous complement inhibitors. Other inhibitors such as C4b-binding protein and factor H, as well as the viral homologue vaccinia virus complement control protein are known to bind heparin and, for the two latter, also to glycosaminoglycans at the surface of cells. We report here that KCP also binds to heparin at physiological ionic strength. With help of site directed mutagenesis, positively charged amino acids in the two N-terminal complement control protein (CCP) domains 1-2 were found to be necessary for heparin binding. In silico molecular docking of heparin to KCP confirmed the experimental data, and further explored the heparin binding site, enabling us to present a model of the KCP-heparin interaction. Furthermore, the docking analysis also yielded insights of the KCP structure, by indicating that the angle between CCP domains 1-2 during the initial binding of heparin is more extended than in the model we have previously presented. We also found that KCP binds to heparan sulfate and weakly to glycosaminoglycans at the surface of cells. This might indicate that KCP at the surface of viral particles aids in the primary attachment to the target cells, which is known to involve binding to heparan sulfate. Therefore, the present study contributes to the knowledge of heparin-protein interactions in general as well as to the understanding of the biology of KSHV.


Asunto(s)
Aminoácidos Básicos/metabolismo , Membrana Celular/metabolismo , Heparina/metabolismo , Proteínas Virales/metabolismo , Aminoácidos Básicos/genética , Animales , Células CHO , Membrana Celular/química , Proteína de Unión al Complemento C4b/antagonistas & inhibidores , Proteína de Unión al Complemento C4b/metabolismo , Cricetinae , Cricetulus , Glicosaminoglicanos/análisis , Glicosaminoglicanos/metabolismo , Heparina/farmacología , Heparitina Sulfato/metabolismo , Humanos , Mutagénesis Sitio-Dirigida , Mutación , Estructura Terciaria de Proteína , Proteínas Virales/genética
9.
Curr Opin Immunol ; 21(3): 266-73, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19502021

RESUMEN

Follicular helper T (T(FH)) cells are a class of helper T cells specialized in the cognate control of antigen-specific B cell immunity. Upon first contact with antigen-primed B cells, pregerminal center effector T(FH) cells promote B cell clonal expansion, antibody isotype switch, plasma cell differentiation, and the induction of germinal centers. By contrast, within germinal centers, T(FH) cells regulate the fate of antigen-specific GC B cells expressing high-affinity variant B cell receptors to promote memory B cell and long-lived plasma cell development. Recent studies unravel multiple signals controlling T(FH) development and functional subtypes of antigen-specific T(FH) cells, including memory T(FH) cells that accelerate memory B cell responses to antigen rechallenge in vivo.


Asunto(s)
Linfocitos B/inmunología , Inmunidad Celular/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Centro Germinal/citología , Centro Germinal/inmunología , Humanos , Memoria Inmunológica/inmunología , Activación de Linfocitos/inmunología , Modelos Inmunológicos
10.
J Biol Chem ; 284(1): 505-514, 2009 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-18990693

RESUMEN

Rhesus rhadinovirus (RRV) is currently the closest known, fully sequenced homolog of human Kaposi sarcoma-associated herpesvirus. Both these viruses encode complement inhibitors as follows: Kaposi sarcoma-associated herpesvirus-complement control protein (KCP) and RRV-complement control protein (RCP). Previously we characterized in detail the functional properties of KCP as a complement inhibitor. Here, we performed comparative analyses for two variants of RCP protein, encoded by RRV strains H26-95 and 17577. Both RCP variants and KCP inhibited human and rhesus complement when tested in hemolytic assays measuring all steps of activation via the classical and the alternative pathway. RCP variants from both RRV strains supported C3b and C4b degradation by factor I and decay acceleration of the classical C3 convertase, similar to KCP. Additionally, the 17577 RCP variant accelerated decay of the alternative C3 convertase, which was not seen for KCP. In contrast to KCP, RCP showed no affinity to heparin and is the first described complement inhibitor in which the binding site for C3b/C4b does not interact with heparin. Molecular modeling shows a structural disruption in the region of RCP that corresponds to the KCP-heparin-binding site. This makes RRV a superior model for future in vivo investigations of complement evasion, as RCP does not play a supportive role in viral attachment as KCP does.


Asunto(s)
Proteínas Inactivadoras de Complemento/metabolismo , Proteínas del Sistema Complemento/metabolismo , Herpesvirus Saimiriino 2/metabolismo , Glicoproteínas de Membrana/metabolismo , Modelos Moleculares , Proteínas Virales/metabolismo , Animales , Sitios de Unión , Células CHO , Proteínas Inactivadoras de Complemento/química , Proteínas Inactivadoras de Complemento/genética , Proteínas del Sistema Complemento/química , Proteínas del Sistema Complemento/genética , Cricetinae , Cricetulus , Heparina/química , Heparina/metabolismo , Herpesvirus Saimiriino 2/química , Herpesvirus Saimiriino 2/genética , Herpesvirus Humano 8/química , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Macaca mulatta , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Especificidad de la Especie , Relación Estructura-Actividad , Proteínas Virales/química , Proteínas Virales/genética , Acoplamiento Viral
11.
J Virol ; 81(8): 4166-76, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17287274

RESUMEN

The diversity of viral strategies to modulate complement activation indicates that this component of the immune system has significant antiviral potential. One example is the Kaposi's sarcoma-associated herpesvirus (KSHV) complement control protein (KCP), which inhibits progression of the complement cascade. Rhesus rhadinovirus (RRV), like KSHV, is a member of the subfamily Gammaherpesvirinae and currently provides the only in vivo model of KSHV pathobiology in primates. In the present study, we characterized the KCP homologue encoded by RRV, RRV complement control protein (RCP). Two strains of RRV have been sequenced to date (H26-95 and 17577), and the RCPs they encode differ substantially in structure: RCP from strain H26-95 has four complement control protein (CCP) domains, whereas RCP from strain 17577 has eight CCP domains. Transcriptional analyses of the RCP gene (ORF4, referred to herein as RCP) in infected rhesus macaque fibroblasts mapped the ends of the transcripts of both strains. They revealed that H26-95 encodes a full-length, unspliced RCP transcript, while 17577 RCP generates a full-length unspliced mRNA and two alternatively spliced transcripts. Western blotting confirmed that infected cells express RCP, and immune electron microscopy disclosed this protein on the surface of RRV virions. Functional studies of RCP encoded by both RRV strains revealed their ability to suppress complement activation by the classical (antibody-mediated) pathway. These data provide the foundation for studies into the biological significance of gammaherpesvirus complement regulatory proteins in a tractable, non-human primate model.


Asunto(s)
Genes Virales , Rhadinovirus/química , Rhadinovirus/genética , Proteínas Virales/genética , Proteínas Virales/fisiología , Animales , Secuencia de Bases , Células CHO , Células Cultivadas , Activación de Complemento , Vía Clásica del Complemento , Cricetinae , Cricetulus , Fibroblastos/virología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Humanos , Macaca mulatta , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Estructura Terciaria de Proteína , Empalme del ARN , ARN Mensajero/biosíntesis , ARN Viral/biosíntesis , Virión/química
12.
J Biol Chem ; 281(6): 3690-7, 2006 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-16330538

RESUMEN

Human C4b-binding protein (C4BP) protects host tissue, and those pathogens able to hijack this plasma glycoprotein, from complement-mediated destruction. We now show that the first two complement control protein (CCP) modules of the C4BP alpha-chain, plus the four residues connecting them, are necessary and sufficient for binding a bacterial virulence factor, the Streptococcus pyogenes M4 (Arp4) protein. Structure determination by NMR reveals two tightly coupled CCP modules in an elongated arrangement within this region of C4BP. Chemical shift perturbation studies demonstrate that the N-terminal, hypervariable region of M4 binds to a site including strand 1 of CCP module 2. This interaction is accompanied by an intermodular reorientation within C4BP. We thus provide a detailed picture of an interaction whereby a pathogen evades complement.


Asunto(s)
Antígenos Bacterianos/química , Proteínas de la Membrana Bacteriana Externa/química , Proteínas Portadoras/química , Proteína de Unión al Complemento C4b/química , Proteína de Unión al Complemento C4b/fisiología , Secuencia de Aminoácidos , Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Sitios de Unión , Calorimetría , Proteínas Portadoras/metabolismo , ADN Complementario/metabolismo , Dimerización , Humanos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Conformación Proteica , Homología de Secuencia de Aminoácido , Virulencia
13.
J Biol Chem ; 278(11): 9283-9, 2003 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-12645526

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is closely associated with Kaposi's sarcoma and certain B-cell lymphomas. The fourth open reading frame of the KSHV genome encodes a protein (KSHV complement control protein (KCP, previously termed ORF4)) predicted to have complement-regulating activity. Here, we show that soluble KCP strongly enhanced the decay of classical C3-convertase but not the alternative pathway C3-convertase, when compared with the host complement regulators: factor H, C4b-binding protein, and decay-accelerating factor. The equilibrium affinity constant (KD) of KCP for C3b and C4b was determined by surface plasmon resonance analysis to range between 0.47-10 microM and 0.025-6.1 microM, respectively, depending on NaCl concentration and cation presence. Soluble and cell-associated KCP acted as a cofactor for factor I (FI)-mediated cleavage of both C4b and C3b and induced the cleavage products C4d and iC3b, respectively. In the presence of KCP, FI further cleaved iC3b to C3d, which has never been described before as complement receptor 1 only mediates the production of C3dg by FI. KCP would enhance virus pathogenesis through evading complement attack, opsonization, and anaphylaxis but may also aid in targeting KSHV to one of its host reservoirs since C3d is a ligand for complement receptor 2 on B-cells.


Asunto(s)
Activación de Complemento , Herpesvirus Humano 8/genética , Proteínas Virales/química , Proteínas Virales/metabolismo , Empalme Alternativo , Complemento C3b/química , Complemento C4b/química , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Humanos , Cinética , Ligandos , Sistemas de Lectura Abierta , Unión Proteica , Estructura Terciaria de Proteína , Resonancia por Plasmón de Superficie
14.
J Biol Chem ; 279(43): 45093-101, 2004 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-15304516

RESUMEN

Kaposi's sarcoma-associated human herpesvirus (KSHV) is thought to cause Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease. Previously, we reported that the KSHV complement control protein (KCP) encoded within the viral genome is a potent regulator of the complement system; it acts both as a cofactor for factor I and accelerates decay of the C3 convertases (Spiller, O. B., Blackbourn, D. J., Mark, L., Proctor, D. G., and Blom, A. M. (2003) J. Biol. Chem. 278, 9283-9289). KCP is a homologue to human complement regulators, being comprised of four complement control protein (CCP) domains. In this, the first study to identify the functional sites of a viral homologue at the amino acid level, we created a three-dimensional homology-based model followed by site-directed mutagenesis to locate complement regulatory sites. Classical pathway regulation, both through decay acceleration and factor I cleavage of C4b, required a cluster of positively charged amino acids in CCP1 stretching into CCP2 (Arg-20, Arg-33, Arg-35, Lys-64, Lys-65, and Lys-88) as well as positively (Lys-131, Lys-133, and His-135) and negatively (Glu-99, Glu-152, and Asp-155) charged areas at opposing faces of the border region between CCPs 2 and 3. The regulation of the alternative pathway (via factor I-mediated C3b cleavage) was found to both overlap with classical pathway regulatory sites (Lys-64, Lys-65, Lys-88 and Lys-131, Lys-133, His-135) as well as require unique, more C-terminal residues in CCPs 3 and 4 (His-158, His-171, and His-213) and CCP 4 (Phe-195, Phe-207, and Leu-209). We show here that KCP has evolved to maintain the spatial structure of its functional sites, especially the positively charged patches, compared with host complement regulators.


Asunto(s)
Proteínas del Sistema Complemento/química , Proteínas del Sistema Complemento/metabolismo , Herpesvirus Humano 8/metabolismo , Proteínas Virales/fisiología , Secuencias de Aminoácidos , Animales , Células CHO , Cromatografía , Dicroismo Circular , Complemento C3b/química , Complemento C4b/química , Cricetinae , Cartilla de ADN/química , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Histidina/química , Humanos , Cinética , Leucina/química , Lisina/química , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Mutación , Fenilalanina/química , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Sefarosa/química , Resonancia por Plasmón de Superficie , Proteínas Virales/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA