Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 13(1): 13668, 2023 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-37608223

RESUMEN

Coronaviruses have been the causative agent of three epidemics and pandemics in the past two decades, including the ongoing COVID-19 pandemic. A broadly-neutralizing coronavirus therapeutic is desirable not only to prevent and treat COVID-19, but also to provide protection for high-risk populations against future emergent coronaviruses. As all coronaviruses use spike proteins on the viral surface to enter the host cells, and these spike proteins share sequence and structural homology, we set out to discover cross-reactive biologic agents targeting the spike protein to block viral entry. Through llama immunization campaigns, we have identified single domain antibodies (VHHs) that are cross-reactive against multiple emergent coronaviruses (SARS-CoV, SARS-CoV-2, and MERS). Importantly, a number of these antibodies show sub-nanomolar potency towards all SARS-like viruses including emergent CoV-2 variants. We identified nine distinct epitopes on the spike protein targeted by these VHHs. Further, by engineering VHHs targeting distinct, conserved epitopes into multi-valent formats, we significantly enhanced their neutralization potencies compared to the corresponding VHH cocktails. We believe this approach is ideally suited to address both emerging SARS-CoV-2 variants during the current pandemic as well as potential future pandemics caused by SARS-like coronaviruses.


Asunto(s)
COVID-19 , Camélidos del Nuevo Mundo , Anticuerpos de Dominio Único , Humanos , Animales , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Pandemias , Epítopos
2.
Front Immunol ; 13: 864775, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35603164

RESUMEN

The SARS-CoV-2 pandemic and particularly the emerging variants have deepened the need for widely available therapeutic options. We have demonstrated that hexamer-enhancing mutations in the Fc region of anti-SARS-CoV IgG antibodies lead to a noticeable improvement in IC50 in both pseudo and live virus neutralization assay compared to parental molecules. We also show that hexamer-enhancing mutants improve C1q binding to target surface. To our knowledge, this is the first time this format has been explored for application in viral neutralization and the studies provide proof-of-concept for the use of hexamer-enhanced IgG1 molecules as potential anti-viral therapeutics.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Inmunoglobulina G/genética , Pruebas Inmunológicas , Pandemias , SARS-CoV-2/genética
3.
Nat Microbiol ; 3(2): 172-180, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29292384

RESUMEN

Epstein-Barr virus (EBV) is an oncogenic virus that infects more than 90% of the world's population 1 . EBV predominantly infects human B cells and epithelial cells, which is initiated by fusion of the viral envelope with a host cellular membrane 2 . The mechanism of EBV entry into B cells has been well characterized 3 . However, the mechanism for epithelial cell entry remains elusive. Here, we show that the integrins αvß5, αvß6 and αvß8 do not function as entry and fusion receptors for epithelial cells, whereas Ephrin receptor tyrosine kinase A2 (EphA2) functions well for both. EphA2 overexpression significantly increased EBV infection of HEK293 cells. Using a virus-free cell-cell fusion assay, we found that EphA2 dramatically promoted EBV but not herpes simplex virus (HSV) fusion with HEK293 cells. EphA2 silencing using small hairpin RNA (shRNA) or knockout by CRISPR-Cas9 blocked fusion with epithelial cells. This inhibitory effect was rescued by the expression of EphA2. Antibody against EphA2 blocked epithelial cell infection. Using label-free surface plasmon resonance binding studies, we confirmed that EphA2 but not EphA4 specifically bound to EBV gHgL and this interaction is through the EphA2 extracellular domain (ECD). The discovery of EphA2 as an EBV epithelial cell receptor has important implications for EBV pathogenesis and may uncover new potential targets that can be used for the development of novel intervention strategies.


Asunto(s)
Efrina-A2/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/virología , Herpesvirus Humano 4/fisiología , Herpesvirus Humano 4/patogenicidad , Internalización del Virus , Animales , Antígenos de Neoplasias/metabolismo , Linfocitos B/virología , Células CHO , Fusión Celular , Cricetulus , Efrina-A2/genética , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Integrinas/metabolismo , ARN Interferente Pequeño , Receptor EphA2 , Receptor EphA4 , Receptores de Vitronectina/metabolismo
4.
Proc Natl Acad Sci U S A ; 114(41): E8703-E8710, 2017 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-28939750

RESUMEN

Herpesvirus entry into cells requires the coordinated action of multiple virus envelope glycoproteins, including gH, gL, and gB. For EBV, the gp42 protein assembles into complexes with gHgL heterodimers and binds HLA class II to activate gB-mediated membrane fusion with B cells. EBV tropism is dictated by gp42 levels in the virion, as it inhibits entry into epithelial cells while promoting entry into B cells. The gHgL and gB proteins are targets of neutralizing antibodies and potential candidates for subunit vaccine development, but our understanding of their neutralizing epitopes and the mechanisms of inhibition remain relatively unexplored. Here we studied the structures and mechanisms of two anti-gHgL antibodies, CL40 and CL59, that block membrane fusion with both B cells and epithelial cells. We determined the structures of the CL40 and CL59 complexes with gHgL using X-ray crystallography and EM to identify their epitope locations. CL59 binds to the C-terminal domain IV of gH, while CL40 binds to a site occupied by the gp42 receptor binding domain. CL40 binding to gHgL/gp42 complexes is not blocked by gp42 and does not interfere with gp42 binding to HLA class II, indicating that its ability to block membrane fusion with B cells represents a defect in gB activation. These data indicate that anti-gHgL neutralizing antibodies can block gHgL-mediated activation of gB through different surface epitopes and mechanisms.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Linfocitos B/virología , Infecciones por Virus de Epstein-Barr/metabolismo , Fusión de Membrana , Glicoproteínas de Membrana/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales/metabolismo , Linfocitos B/citología , Linfocitos B/inmunología , Células Cultivadas , Células Epiteliales/citología , Células Epiteliales/inmunología , Células Epiteliales/virología , Epítopos , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/patogenicidad , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/inmunología , Chaperonas Moleculares/química , Chaperonas Moleculares/inmunología , Unión Proteica , Conformación Proteica , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/inmunología , Proteínas Virales/química , Proteínas Virales/inmunología , Internalización del Virus
5.
J Biol Chem ; 292(43): 17626-17642, 2017 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-28887305

RESUMEN

In many Gram-negative bacteria, the peptidoglycan synthase PBP1A requires the outer membrane lipoprotein LpoA for constructing a functional peptidoglycan required for bacterial viability. Previously, we have shown that the C-terminal domain of Haemophilus influenzae LpoA (HiLpoA) has a highly conserved, putative substrate-binding cleft between two α/ß lobes. Here, we report a 2.0 Å resolution crystal structure of the HiLpoA N-terminal domain. Two subdomains contain tetratricopeptide-like motifs that form a concave groove, but their relative orientation differs by ∼45° from that observed in an NMR structure of the Escherichia coli LpoA N domain. We also determined three 2.0-2.8 Å resolution crystal structures containing four independent full-length HiLpoA molecules. In contrast to an elongated model previously suggested for E. coli LpoA, each HiLpoA formed a U-shaped structure with a different C-domain orientation. This resulted from both N-domain twisting and rotation of the C domain (up to 30°) at the end of the relatively immobile interdomain linker. Moreover, a previously predicted hinge between the lobes of the LpoA C domain exhibited variations of up to 12°. Small-angle X-ray scattering data revealed excellent agreement with a model calculated by normal mode analysis from one of the full-length HiLpoA molecules but even better agreement with an ensemble of this molecule and two of the partially extended normal mode analysis-predicted models. The different LpoA structures helped explain how an outer membrane-anchored LpoA can either withdraw from or extend toward the inner membrane-bound PBP1A through peptidoglycan gaps and hence regulate the synthesis of peptidoglycan necessary for bacterial viability.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/química , Activadores de Enzimas/química , Haemophilus influenzae/química , Proteínas de Unión a las Penicilinas , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Cristalografía por Rayos X , Activadores de Enzimas/metabolismo , Haemophilus influenzae/genética , Haemophilus influenzae/metabolismo , Resonancia Magnética Nuclear Biomolecular , Dominios Proteicos
6.
Curr Opin Virol ; 24: 97-104, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28538165

RESUMEN

Enveloped viruses have evolved diverse transmembrane proteins and protein complexes to enable host cell entry by regulating and activating membrane fusion in a target cell-specific manner. In general terms, the entry process requires a receptor binding step, an activation step and a membrane fusion step, which can be encoded within a single viral protein or distributed among multiple viral proteins. HIV and influenza virus, for example, encode all of these functions in a single trimeric glycoprotein, HIV env or influenza virus hemagglutinin (HA). In contrast, herpesviruses have the host receptor binding, activation and fusogenic roles distributed among multiple envelope glycoproteins (ranging from three to six), which must coordinate their functions at the site of fusion. Despite the apparent complexity in the number of viral entry proteins, herpesvirus entry is fundamentally built around two core glycoprotein entities: the gHgL complex, which appears to act as an 'activator' of entry, and the gB protein, which is thought to act as the membrane 'fusogen'. Both are required for all herpesvirus fusion and entry. In many herpesviruses, gHgL either binds host receptors directly or assembles into larger complexes with additional viral proteins that bind host receptors, conferring specificity to the cells that are targeted for infection. These gHgL entry complexes (ECs) are centrally important to activating gB-mediated membrane fusion and establishing viral tropism, forming membrane bridging intermediates before gB triggering. Here we review recent structural and functional studies of Epstein-Barr virus (EBV) and Cytomegalovirus (CMV) gHgL complexes that provide a framework for understanding the role of gHgL in herpesvirus entry. Furthermore, a recently determined EM model of Herpes Simplex virus (HSV) gB embedded in exosomes highlights how gB conformational changes may promote viral and cellular membrane fusion.


Asunto(s)
Herpesviridae/fisiología , Tropismo Viral , Internalización del Virus , Citomegalovirus/fisiología , Exosomas/química , Exosomas/fisiología , Hemaglutininas/metabolismo , Herpesvirus Humano 4/fisiología , Interacciones Huésped-Patógeno , Humanos , Simplexvirus/química , Simplexvirus/fisiología , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales de Fusión/metabolismo , Proteínas Virales/metabolismo
7.
Nat Commun ; 7: 13557, 2016 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-27929061

RESUMEN

Herpesvirus entry into host cells is mediated by multiple virally encoded receptor binding and membrane fusion glycoproteins. Despite their importance in host cell tropism and associated disease pathology, the underlying and essential interactions between these viral glycoproteins remain poorly understood. For Epstein-Barr virus (EBV), gHgL/gp42 complexes bind HLA class II to activate membrane fusion with B cells, but gp42 inhibits fusion and entry into epithelial cells. To clarify the mechanism by which gp42 controls the cell specificity of EBV infection, here we determined the structure of gHgL/gp42 complex bound to an anti-gHgL antibody (E1D1). The critical regulator of EBV tropism is the gp42 N-terminal domain, which tethers the HLA-binding domain to gHgL by wrapping around the exterior of three gH domains. Both the gp42 N-terminal domain and E1D1 selectively inhibit epithelial-cell fusion; however, they engage distinct surfaces of gHgL. These observations clarify key determinants of EBV host cell tropism.


Asunto(s)
Herpesvirus Humano 4/fisiología , Proteínas del Envoltorio Viral/fisiología , Tropismo Viral , Animales , Células CHO , Cricetulus , Células Epiteliales/virología , Herpesvirus Humano 4/química , Mutación , Conformación Proteica , Proteínas del Envoltorio Viral/química
8.
Nat Commun ; 7: 11610, 2016 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-27194387

RESUMEN

Omalizumab is a widely used therapeutic anti-IgE antibody. Here we report the crystal structure of the omalizumab-Fab in complex with an IgE-Fc fragment. This structure reveals the mechanism of omalizumab-mediated inhibition of IgE interactions with both high- and low-affinity IgE receptors, and explains why omalizumab selectively binds free IgE. The structure of the complex also provides mechanistic insight into a class of disruptive IgE inhibitors that accelerate the dissociation of the high-affinity IgE receptor from IgE. We use this structural data to generate a mutant IgE-Fc fragment that is resistant to omalizumab binding. Treatment with this omalizumab-resistant IgE-Fc fragment, in combination with omalizumab, promotes the exchange of cell-bound full-length IgE with omalizumab-resistant IgE-Fc fragments on human basophils. This combination treatment also blocks basophil activation more efficiently than either agent alone, providing a novel approach to probe regulatory mechanisms underlying IgE hypersensitivity with implications for therapeutic interventions.


Asunto(s)
Antialérgicos/farmacología , Inmunoglobulina E/efectos de los fármacos , Omalizumab/farmacología , Receptores de IgE/antagonistas & inhibidores , Animales , Antialérgicos/química , Basófilos/efectos de los fármacos , Línea Celular , Sinergismo Farmacológico , Humanos , Inmunoglobulina E/química , Inmunoglobulina E/genética , Mutación , Omalizumab/química , Conformación Proteica
9.
Mol Cells ; 39(4): 286-91, 2016 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-27094060

RESUMEN

Epstein-Barr virus (EBV) is the prototypical γ-herpesvirus and an obligate human pathogen that infects mainly epithelial cells and B cells, which can result in malignancies. EBV infects these target cells by fusing with the viral and cellular lipid bilayer membranes using multiple viral factors and host receptor(s) thus exhibiting a unique complexity in its entry machinery. To enter epithelial cells, EBV requires minimally the conserved core fusion machinery comprised of the glycoproteins gH/gL acting as the receptor-binding complex and gB as the fusogen. EBV can enter B cells using gp42, which binds tightly to gH/gL and interacts with host HLA class II, activating fusion. Previously, we published the individual crystal structures of EBV entry factors, such as gH/gL and gp42, the EBV/host receptor complex, gp42/HLA-DR1, and the fusion protein EBV gB in a postfusion conformation, which allowed us to identify structural determinants and regions critical for receptor-binding and membrane fusion. Recently, we reported different low resolution models of the EBV B cell entry triggering complex (gHgL/gp42/HLA class II) in "open" and "closed" states based on negative-stain single particle electron microscopy, which provide further mechanistic insights. This review summarizes the current knowledge of these key players in EBV entry and how their structures impact receptor-binding and the triggering of gB-mediated fusion.


Asunto(s)
Herpesvirus Humano 4/fisiología , Antígenos de Histocompatibilidad Clase II/metabolismo , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/metabolismo , Linfocitos B/virología , Línea Celular , Células Epiteliales/virología , Genes MHC Clase II , Antígenos de Histocompatibilidad Clase II/química , Humanos , Modelos Moleculares , Estructura Terciaria de Proteína , Tropismo Viral , Internalización del Virus
10.
J Virol ; 88(23): 13570-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25231307

RESUMEN

UNLABELLED: Epstein-Barr virus (EBV) infects target cells via fusion with cellular membranes. For entry into epithelial cells, EBV requires the herpesvirus conserved core fusion machinery, composed of glycoprotein B (gB) and gH/gL. In contrast, for B cell fusion it requires gB and gH/gL with gp42 serving as a cell tropism switch. The available crystal structures for gH/gL allow the targeted analysis of structural determinants of gH to identify functional regions critical for membrane fusion. Domain II of EBV gH contains two disulfide bonds (DBs). The first is unique for EBV and closely related gammaherpesviruses. The second is conserved across the beta- and gammaherpesviruses and is positioned to stabilize a putative syntaxin-like bundle motif. To analyze the role of these DBs in membrane fusion, gH was mutated by amino acid substitution of the DB cysteines. Mutation of the EBV-specific DB resulted in diminished gH/gL cell surface expression that correlated with diminished B cell and epithelial cell fusion. In contrast, mutation of the conserved DB resulted in wild-type-like B cell fusion, whereas epithelial cell fusion was greatly reduced. The gH mutants bound well to gp42 but had diminished binding to epithelial cells. Tyrosine 336, located adjacent to cysteine 335 of the conserved DB, also was found to be important for DB stabilization and gH/gL function. We conclude that the conserved DB has a cell type-specific function, since it is important for the binding of gH to epithelial cells initiating epithelial cell fusion but not for fusion with B cells and gp42 binding. IMPORTANCE: EBV predominantly infects epithelial and B cells in humans, which can result in EBV-associated cancers, such as Burkitt and Hodgkin lymphoma, as well as nasopharyngeal carcinoma. EBV is also associated with a variety of lymphoproliferative disorders, typically of B cell origin, observed in immunosuppressed individuals, such as posttransplant or HIV/AIDS patients. The gH/gL complex plays an essential but still poorly characterized role as an important determinant for EBV cell tropism. In the current studies, we found that mutants in the DB C278/C335 and the neighboring tyrosine 336 have cell type-specific functional deficits with selective decreases in epithelial cell, but not B cell, binding and fusion. The present study brings new insights into the gH function as a determinant for epithelial cell tropism during herpesvirus-induced membrane fusion and highlights a specific gH motif required for epithelial cell fusion.


Asunto(s)
Linfocitos B/virología , Disulfuros/metabolismo , Células Epiteliales/virología , Herpesvirus Humano 4/fisiología , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Animales , Línea Celular , Análisis Mutacional de ADN , Herpesvirus Humano 4/genética , Humanos , Modelos Moleculares , Conformación Proteica , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética
11.
PLoS Pathog ; 10(8): e1004309, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25144748

RESUMEN

Epstein-Barr Virus (EBV) is an enveloped double-stranded DNA virus of the gammaherpesvirinae sub-family that predominantly infects humans through epithelial cells and B cells. Three EBV glycoproteins, gH, gL and gp42, form a complex that targets EBV infection of B cells. Human leukocyte antigen (HLA) class II molecules expressed on B cells serve as the receptor for gp42, triggering membrane fusion and virus entry. The mechanistic role of gHgL in herpesvirus entry has been largely unresolved, but it is thought to regulate the activation of the virally-encoded gB protein, which acts as the primary fusogen. Here we study the assembly and function of the reconstituted B cell entry complex comprised of gHgL, gp42 and HLA class II. The structure from negative-stain electron microscopy provides a detailed snapshot of an intermediate state in EBV entry and highlights the potential for the triggering complex to bring the two membrane bilayers into proximity. Furthermore, gHgL interacts with a previously identified, functionally important hydrophobic pocket on gp42, defining the overall architecture of the complex and playing a critical role in membrane fusion activation. We propose a macroscopic model of the initiating events in EBV B cell fusion centered on the formation of the triggering complex in the context of both viral and host membranes. This model suggests how the triggering complex may bridge the two membrane bilayers, orienting critical regions of the N- and C- terminal ends of gHgL to promote the activation of gB and efficient membrane fusion.


Asunto(s)
Linfocitos B/virología , Infecciones por Virus de Epstein-Barr/metabolismo , Herpesvirus Humano 4/patogenicidad , Interacciones Huésped-Parásitos/fisiología , Internalización del Virus , Animales , Células CHO , Cricetinae , Cricetulus , Antígenos HLA-DQ/metabolismo , Procesamiento de Imagen Asistido por Computador , Glicoproteínas de Membrana/metabolismo , Microscopía Electrónica , Chaperonas Moleculares/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales/metabolismo
12.
PLoS One ; 7(6): e37984, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22675501

RESUMEN

Capsules frequently play a key role in bacterial interactions with their environment. Escherichia coli capsules were categorized as groups 1 through 4, each produced by a distinct mechanism. Etk and Etp are members of protein families required for the production of group 1 and group 4 capsules. These members function as a protein tyrosine kinase and protein tyrosine phosphatase, respectively. We show that Etp dephosphorylates Etk in vivo, and mutations rendering Etk or Etp catalytically inactive result in loss of group 4 capsule production, supporting the notion that cyclic phosphorylation and dephosphorylation of Etk is required for capsule formation. Notably, Etp also becomes tyrosine phosphorylated in vivo and catalyzes rapid auto-dephosphorylation. Further analysis identified Tyr121 as the phosphorylated residue of Etp. Etp containing Phe, Glu or Ala in place of Tyr121 retained phosphatase activity and catalyzed dephosphorylation of Etp and Etk. Although EtpY121E and EtpY121A still supported capsule formation, EtpY121F failed to do so. These results suggest that cycles of phosphorylation and dephosphorylation of Etp, as well as Etk, are involved in the formation of group 4 capsule, providing an additional regulatory layer to the complex control of capsule production.


Asunto(s)
Cápsulas Bacterianas/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Proteínas de la Membrana/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Secuencia de Aminoácidos , Cinética , Modelos Biológicos , Datos de Secuencia Molecular , Mutación/genética , Fosforilación , Fosfotirosina/metabolismo , Espectrometría de Masa por Ionización de Electrospray
13.
Biochemistry ; 50(24): 5465-76, 2011 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-21449614

RESUMEN

We report the 1.9 Å resolution crystal structure of enteropathogenic Escherichia coli GfcC, a periplasmic protein encoded by the gfc operon, which is essential for assembly of group 4 polysaccharide capsule (O-antigen capsule). Presumed gene orthologs of gfcC are present in capsule-encoding regions of at least 29 genera of Gram-negative bacteria. GfcC, a member of the DUF1017 family, is comprised of tandem ß-grasp (ubiquitin-like) domains (D2 and D3) and a carboxyl-terminal amphipathic helix, a domain arrangement reminiscent of that of Wza that forms an exit pore for group 1 capsule export. Unlike the membrane-spanning C-terminal helix from Wza, the GfcC C-terminal helix packs against D3. Previously unobserved in a ß-grasp domain structure is a 48-residue helical hairpin insert in D2 that binds to D3, constraining its position and sequestering the carboxyl-terminal amphipathic helix. A centrally located and invariant Arg115 not only is essential for proper localization but also forms one of two mostly conserved pockets. Finally, we draw analogies between a GfcC protein fused to an outer membrane ß-barrel pore in some species and fusion proteins necessary for secreting biofilm-forming exopolysaccharides.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/química , Proteínas de Escherichia coli/química , Secuencia de Aminoácidos , Cápsulas Bacterianas/química , Proteínas de la Membrana Bacteriana Externa/genética , Secuencia de Bases , Secuencia Conservada , Cristalografía por Rayos X , ADN Bacteriano/genética , Dimerización , Escherichia coli Enteropatógena/química , Escherichia coli Enteropatógena/genética , Proteínas de Escherichia coli/genética , Genes Bacterianos , Modelos Moleculares , Datos de Secuencia Molecular , Operón , Dominios y Motivos de Interacción de Proteínas , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Homología de Secuencia de Aminoácido , Electricidad Estática
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA