Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 12(1): 13413, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35927444

RESUMEN

While vaccination against HIV-1 has been so far unsuccessful, recently broadly neutralizing antibodies (bNAbs) against HIV-1 envelope glycoprotein were shown to induce long-term suppression in the absence of antiretroviral therapy in patients with antibody-sensitive viral reservoirs. The requirement of neutralizing antibodies indicates that the antibody mediated removal (clearance) of HIV-1 in itself is not efficient enough in these immune compromised patients. Here we present a novel, alternative approach that is independent of a functional immune system to clear HIV-1, by capturing the virus and redirecting it to non-target cells where it is internalized and degraded. We use bispecific antibodies with domains derived from small single chain Llama antibodies (VHHs). These bind with one domain to HIV-1 envelope proteins and with the other domain direct the virus to cells expressing epidermal growth factor receptor (EGFR), a receptor that is ubiquitously expressed in the body. We show that HIV envelope proteins, virus-like particles and HIV-1 viruses (representing HIV-1 subtypes A, B and C) are efficiently recruited to EGFR, internalized and degraded in the lysosomal pathway at low nM concentrations of bispecific VHHs. This directed degradation in non-target cells may provide a clearance platform for the removal of viruses and other unwanted agents from the circulation, including toxins, and may thus provide a novel method for curing.


Asunto(s)
Camélidos del Nuevo Mundo , Infecciones por VIH , Seropositividad para VIH , VIH-1 , Anticuerpos de Cadena Única , Animales , Anticuerpos Neutralizantes , Receptores ErbB , Anticuerpos Anti-VIH , Humanos
2.
Protein Eng Des Sel ; 29(4): 123-33, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26945588

RESUMEN

Highly potent human antibodies are required to therapeutically neutralize cytokines such as interleukin-6 (IL-6) that is involved in many inflammatory diseases and malignancies. Although a number of mutagenesis approaches exist to perform antibody affinity maturation, these may cause antibody instability and production issues. Thus, a robust and easy antibody affinity maturation strategy to increase antibody potency remains highly desirable. By immunizing llama, cloning the 'immune' antibody repertoire and using phage display, we selected a diverse set of IL-6 antagonistic Fabs. Heavy chain shuffling was performed on the Fab with lowest off-rate, resulting in a panel of variants with even lower off-rate. Structural analysis of the Fab:IL-6 complex suggests that the increased affinity was partly due to a serine to tyrosine switch in HCDR2. This translated into neutralizing capacity in an in vivo model of IL-6 induced SAA production. Finally, a novel Fab library was designed, encoding all variations found in the natural repertoire of VH genes identified after heavy chain shuffling. High stringency selections resulted in identification of a Fab with 250-fold increased potency when re-formatted into IgG1. Compared with a heavily engineered anti-IL-6 monoclonal antibody currently in clinical development, this IgG was at least equally potent, showing the engineering process to have had led to a highly potent anti-IL-6 antibody.


Asunto(s)
Fragmentos Fab de Inmunoglobulinas/genética , Fragmentos Fab de Inmunoglobulinas/metabolismo , Mutación/genética , Biblioteca de Péptidos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Secuencia de Aminoácidos , Animales , Afinidad de Anticuerpos , Camélidos del Nuevo Mundo/genética , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Interleucina-6/inmunología , Modelos Inmunológicos , Modelos Moleculares , Proteínas Recombinantes/química , Alineación de Secuencia
3.
MAbs ; 7(4): 693-706, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26018625

RESUMEN

Camelid immunoglobulin variable (IGV) regions were found homologous to their human counterparts; however, the germline V repertoires of camelid heavy and light chains are still incomplete and their therapeutic potential is only beginning to be appreciated. We therefore leveraged the publicly available HTG and WGS databases of Lama pacos and Camelus ferus to retrieve the germline repertoire of V genes using human IGV genes as reference. In addition, we amplified IGKV and IGLV genes to uncover the V germline repertoire of Lama glama and sequenced BAC clones covering part of the Lama pacos IGK and IGL loci. Our in silico analysis showed that camelid counterparts of all human IGKV and IGLV families and most IGHV families could be identified, based on canonical structure and sequence homology. Interestingly, this sequence homology seemed largely restricted to the Ig V genes and was far less apparent in other genes: 6 therapeutically relevant target genes differed significantly from their human orthologs. This contributed to efficient immunization of llamas with the human proteins CD70, MET, interleukin (IL)-1ß and IL-6, resulting in large panels of functional antibodies. The in silico predicted human-homologous canonical folds of camelid-derived antibodies were confirmed by X-ray crystallography solving the structure of 2 selected camelid anti-CD70 and anti-MET antibodies. These antibodies showed identical fold combinations as found in the corresponding human germline V families, yielding binding site structures closely similar to those occurring in human antibodies. In conclusion, our results indicate that active immunization of camelids can be a powerful therapeutic antibody platform.


Asunto(s)
Región Variable de Inmunoglobulina , Pliegue de Proteína , Homología de Secuencia de Aminoácido , Animales , Camélidos del Nuevo Mundo , Camelus , Cristalografía por Rayos X , Humanos , Región Variable de Inmunoglobulina/química , Región Variable de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/inmunología , Estructura Terciaria de Proteína
4.
MAbs ; 6(2): 523-32, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24492296

RESUMEN

Overexpression of CD70 has been documented in a variety of solid and hematological tumors, where it is thought to play a role in tumor proliferation and evasion of immune surveillance. Here, we describe ARGX-110, a defucosylated IgG1 monoclonal antibody (mAb) that selectively targets and neutralizes CD70, the ligand of CD27.   ARGX-110 was generated by immunization of outbred llamas. The antibody was germlined to 95% human identity, and its anti-tumor efficacy was tested in several in vitro assays. ARGX-110 binds CD70 with picomolar affinity. In depletion studies, ARGX-110 lyses tumor cells with greater efficacy than its fucosylated version. In addition, ARGX-110 demonstrates strong complement-dependent cytotoxicity and antibody-dependent cellular phagocytosis activity. ARGX-110 inhibits signaling of CD27, which results in blocking of the activation and proliferation of Tregs. In a Raji xenograft model, administration of the fucosylated version of ARGX-110 resulted in a prolonged survival at doses of 0.1 mg/kg and above. The pharmacokinetics of ARGX-110 was tested in cynomolgus monkeys; the calculated half-life is 12 days. In conclusion, ARGX-110 is a potent blocking mAb with a dual mode of action against both CD70-bearing tumor cells and CD70-dependent Tregs. This antibody is now in a Phase 1 study in patients with advanced malignancies expressing CD70 (NCT01813539).


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Antineoplásicos/metabolismo , Ligando CD27/inmunología , Inmunoglobulina G/metabolismo , Inmunoterapia/métodos , Neoplasias/terapia , Linfocitos T Reguladores/efectos de los fármacos , Animales , Anticuerpos Monoclonales/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Antineoplásicos/inmunología , Camélidos del Nuevo Mundo , Puntos de Control del Ciclo Celular/inmunología , Células Cultivadas , Humanos , Inmunoglobulina G/inmunología , Activación de Linfocitos/efectos de los fármacos , Neoplasias/inmunología , Transducción de Señal/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/antagonistas & inhibidores
5.
Sci Rep ; 3: 1118, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23346375

RESUMEN

FcRn is a key player in several immunological and non-immunological processes, as it mediates maternal-fetal transfer of IgG, regulates the serum persistence of IgG and albumin, and transports both ligands between different cellular compartments. In addition, FcRn enhances antigen presentation. Thus, there is an intense interest in studies of how FcRn binds and transports its cargo within and across several types of cells, and FcRn detection reagents are in high demand. Here we report on phage display-selected Nanobodies that target human FcRn. The Nanobodies were obtained from a variable-domain repertoire library isolated from a llama immunized with recombinant human FcRn. One candidate, Nb218-H4, was shown to bind FcRn with high affinity at both acidic and neutral pH, without competing ligand binding and interfering with FcRn functions, such as transcytosis of IgG. Thus, Nb218-H4 can be used as a detection probe and as a tracker for visualization of FcRn-mediated cellular transport.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Receptores Fc/inmunología , Receptores Fc/metabolismo , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/metabolismo , Animales , Transporte Biológico , Camélidos del Nuevo Mundo/inmunología , Camélidos del Nuevo Mundo/metabolismo , Células HEK293 , Haplorrinos , Humanos , Concentración de Iones de Hidrógeno , Inmunoglobulina G/inmunología , Ligandos , Ratones , Ratas , Porcinos , Transcitosis/inmunología
6.
PLoS One ; 6(10): e26299, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22022593

RESUMEN

The polymeric immunoglobulin receptor (pIgR) ensures the transport of dimeric immunoglobulin A (dIgA) and pentameric immunoglobulin M (pIgM) across epithelia to the mucosal layer of for example the intestines and the lungs via transcytosis. Per day the human pIgR mediates the excretion of 2 to 5 grams of dIgA into the mucosa of luminal organs. This system could prove useful for therapies aiming at excretion of compounds into the mucosa. Here we investigated the use of the variable domain of camelid derived heavy chain only antibodies, also known as VHHs or Nanobodies®, targeting the human pIgR, as a transport system across epithelial cells. We show that VHHs directed against the human pIgR are able to bind the receptor with high affinity (∼1 nM) and that they compete with the natural ligand, dIgA. In a transcytosis assay both native and phage-bound VHH were only able to get across polarized MDCK cells that express the human pIgR gene in a basolateral to apical fashion. Indicating that the VHHs are able to translocate across epithelia and to take along large particles of cargo. Furthermore, by making multivalent VHHs we were able to enhance the transport of the compounds both in a MDCK-hpIgR and Caco-2 cell system, probably by inducing receptor clustering. These results show that VHHs can be used as a carrier system to exploit the human pIgR transcytotic system and that multivalent compounds are able to significantly enhance the transport across epithelial monolayers.


Asunto(s)
Cadenas Pesadas de Inmunoglobulina/metabolismo , Región Variable de Inmunoglobulina/metabolismo , Receptores de Inmunoglobulina Polimérica/metabolismo , Transcitosis , Animales , Células CACO-2 , Camélidos del Nuevo Mundo , Polaridad Celular , Perros , Células Epiteliales/citología , Células Epiteliales/metabolismo , Mapeo Epitopo , Humanos , Unión Proteica , Reproducibilidad de los Resultados
7.
PLoS One ; 6(4): e17665, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21483777

RESUMEN

For efficient prevention of viral infections and cross protection, simultaneous targeting of multiple viral epitopes is a powerful strategy. Llama heavy chain antibody fragments (VHH) against the trimeric envelope proteins of Respiratory Syncytial Virus (Fusion protein), Rabies virus (Glycoprotein) and H5N1 Influenza (Hemagglutinin 5) were selected from llama derived immune libraries by phage display. Neutralizing VHH recognizing different epitopes in the receptor binding sites on the spikes with affinities in the low nanomolar range were identified for all the three viruses by viral neutralization assays. By fusion of VHH with variable linker lengths, multimeric constructs were made that improved neutralization potencies up to 4,000-fold for RSV, 1,500-fold for Rabies virus and 75-fold for Influenza H5N1. The potencies of the VHH constructs were similar or better than best performing monoclonal antibodies. The cross protection capacity against different viral strains was also improved for all three viruses, both by multivalent (two or three identical VHH) and biparatopic (two different VHH) constructs. By combining a VHH neutralizing RSV subtype A, but not subtype B with a poorly neutralizing VHH with high affinity for subtype B, a biparatopic construct was made with low nanomolar neutralizing potency against both subtypes. Trivalent anti-H5N1 VHH neutralized both Influenza H5N1 clade1 and 2 in a pseudotype assay and was very potent in neutralizing the NIBRG-14 Influenza H5N1 strain with IC(50) of 9 picomolar. Bivalent and biparatopic constructs against Rabies virus cross neutralized both 10 different Genotype 1 strains and Genotype 5.The results show that multimerization of VHH fragments targeting multiple epitopes on a viral trimeric spike protein is a powerful tool for anti-viral therapy to achieve "best-in-class" and broader neutralization capacity.


Asunto(s)
Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Neutralizantes/inmunología , Camélidos del Nuevo Mundo , Cadenas Pesadas de Inmunoglobulina/biosíntesis , Cadenas Pesadas de Inmunoglobulina/inmunología , Virus/inmunología , Animales , Anticuerpos Neutralizantes/aislamiento & purificación , Especificidad de Anticuerpos , Antivirales/inmunología , Antivirales/aislamiento & purificación , Antivirales/metabolismo , Reacciones Cruzadas , Relación Dosis-Respuesta Inmunológica , Epítopos/inmunología , Genotipo , Cadenas Pesadas de Inmunoglobulina/aislamiento & purificación , Subtipo H5N1 del Virus de la Influenza A/inmunología , Lyssavirus/genética , Lyssavirus/inmunología , Virus Sincitiales Respiratorios/inmunología , Proteínas Virales/inmunología
8.
Proc Natl Acad Sci U S A ; 107(47): 20565-70, 2010 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-21059953

RESUMEN

The important family of G protein-coupled receptors has so far not been targeted very successfully with conventional monoclonal antibodies. Here we report the isolation and characterization of functional VHH-based immunoglobulin single variable domains (or nanobodies) against the chemokine receptor CXCR4. Two highly selective monovalent nanobodies, 238D2 and 238D4, were obtained using a time-efficient whole cell immunization, phage display, and counterselection method. The highly selective VHH-based immunoglobulin single variable domains competitively inhibited the CXCR4-mediated signaling and antagonized the chemoattractant effect of the CXCR4 ligand CXCL12. Epitope mapping showed that the two nanobodies bind to distinct but partially overlapping sites in the extracellular loops. Short peptide linkage of 238D2 with 238D4 resulted in significantly increased affinity for CXCR4 and picomolar activity in antichemotactic assays. Interestingly, the monovalent nanobodies behaved as neutral antagonists, whereas the biparatopic nanobodies acted as inverse agonists at the constitutively active CXCR4-N3.35A. The CXCR4 nanobodies displayed strong antiretroviral activity against T cell-tropic and dual-tropic HIV-1 strains. Moreover, the biparatopic nanobody effectively mobilized CD34-positive stem cells in cynomolgus monkeys. Thus, the nanobody platform may be highly effective at generating extremely potent and selective G protein-coupled receptor modulators.


Asunto(s)
Anticuerpos/farmacología , Quimiotaxis/efectos de los fármacos , VIH-1 , Receptores CXCR4/inmunología , Replicación Viral/efectos de los fármacos , Animales , Anticuerpos/aislamiento & purificación , Antígenos CD34 , Bencilaminas , Sitios de Unión/genética , Células COS , Chlorocebus aethiops , Ciclamas , Ensayo de Inmunoadsorción Enzimática , Mapeo Epitopo , Células HEK293 , Movilización de Célula Madre Hematopoyética , Compuestos Heterocíclicos , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
Mol Cancer Ther ; 7(8): 2288-97, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18723476

RESUMEN

The approximately 15-kDa variable domains of camelid heavy-chain-only antibodies (called Nanobodies) can easily be formatted as multivalent or multispecific single-chain proteins. Because of fast excretion, however, they are less suitable for therapy of cancer. In this study, we aimed for improved tumor targeting of a bivalent anti-epidermal growth factor receptor (EGFR) Nanobody (alphaEGFR-alphaEGFR) by fusion to a Nanobody unit binding to albumin (alphaAlb). Biodistributions of alphaEGFR-alphaEGFR, alphaEGFR-alphaEGFR-alphaAlb ( approximately 50 kDa), alphaTNF-alphaTNF-alphaAlb (control, binding tumor necrosis factor-alpha), and the approximately 150-kDa anti-EGFR antibody cetuximab were compared in A431 xenograft-bearing mice. The proteins were radiolabeled with (177)Lu to facilitate quantification. Tumor uptake of (177)Lu-alphaEGFR-alphaEGFR decreased from 5.0 +/- 1.4 to 1.1 +/- 0.1 %ID/g between 6 and 72 h after injection. Due to its rapid blood clearance, tumor-to-blood ratios >80 were obtained within 6 h after injection. Blood clearance became dramatically slower and tumor uptake became significantly higher by introduction of alphaAlb. Blood levels of alphaEGFR-alphaEGFR-alphaAlb were 21.2 +/- 2.5, 11.9 +/- 0.6, and 4.0 +/- 1.4 and tumor levels were 19.4 +/- 5.5, 35.2 +/- 7.5, and 28.0 +/- 6.8 %ID/g at 6, 24, and 72 h after injection, respectively. Tumor uptake was at least as high as for cetuximab (15.5 +/- 3.9, 27.1 +/- 7.9, and 25.6 +/- 6.1 %ID/g) and significantly higher than for alphaTNF-alphaTNF-alphaAlb. alphaEGFR-alphaEGFR-alphaAlb showed faster and deeper tumor penetration than cetuximab. These data show that simple fusion of alphaEGFR and alphaAlb building blocks results in a bifunctional Nanobody format, which seems more favorable for therapy as far as pharmacokinetics and tumor deposition are concerned.


Asunto(s)
Albúminas/metabolismo , Anticuerpos/inmunología , Receptores ErbB/inmunología , Nanotecnología , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales Humanizados , Cetuximab , Receptores ErbB/metabolismo , Unión Proteica , Distribución Tisular
10.
J Immunol Methods ; 324(1-2): 1-12, 2007 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-17570391

RESUMEN

Large scale, highly specific purification of valuable proteins from blood and removal of undesirable components promise to have wide therapeutic applications. Moreover, depletion of bulk proteins from blood is a prerequisite for clinical proteomics. Here we describe the development of specific, high affinity Camelid antibody fragments (VHH) derived from immune libraries for purification and depletion of the bulk protein HSA and IgG from human serum and plasma for therapeutic and research purposes. The anti-IgG VHH substantially improved depletion of IgGs from blood over the classical method based on protein A. To demonstrate the improved performance of VHH based IgG depletion, we analyzed the presence of auto-antibodies in human plasma before and after depletion from two groups of patients with auto-immune disease: Goodpasture syndrome (GP) and systemic lupus erythematosus (SLE). VHHs can be produced efficiently and cost effectively in Saccharomyces cerevisiae, a genetically regarded as safe (GRAS) microorganism. A good manufacturing process (GMP) for purification of these VHHs has also been developed. Moreover, as VHHs are single protein chains, they can be coupled relatively easily to solid matrices. These three factors are important for developing affinity purification medication.


Asunto(s)
Marcadores de Afinidad , Anticuerpos Antiidiotipos/metabolismo , Cromatografía de Afinidad , Inmunoglobulina G/inmunología , Cadenas Pesadas de Inmunoglobulina/metabolismo , Región Variable de Inmunoglobulina/metabolismo , Albúmina Sérica/inmunología , Albúmina Sérica/metabolismo , Animales , Especificidad de Anticuerpos , Sitios de Unión de Anticuerpos , Camélidos del Nuevo Mundo , Humanos , Ligandos , Unión Proteica
11.
Cancer Immunol Immunother ; 56(3): 303-317, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16738850

RESUMEN

The development of a number of different solid tumours is associated with over-expression of ErbB1, or the epidermal growth factor receptor (EGFR), and this over-expression is often correlated with poor prognosis of patients. Therefore, this receptor tyrosine kinase is considered to be an attractive target for antibody-based therapy. Indeed, antibodies to the EGFR have already proven their value for the treatment of several solid tumours, especially in combination with chemotherapeutic treatment regimens. Variable domains of camelid heavy chain-only antibodies (called Nanobodies) have superior properties compared with classical antibodies in that they are small, very stable, easy to produce in large quantities and easy to re-format into multi-valent or multi-specific proteins. Furthermore, they can specifically be selected for a desired function by phage antibody display. In this report, we describe the successful selection and the characterisation of antagonistic anti-EGFR Nanobodies. By using a functional selection strategy, Nanobodies that specifically competed for EGF binding to the EGFR were isolated from "immune" phage Nanobody repertoires. The selected antibody fragments were found to efficiently inhibit EGF binding to the EGFR without acting as receptor agonists themselves. In addition, they blocked EGF-mediated signalling and EGF-induced cell proliferation. In an in vivo murine xenograft model, the Nanobodies were effective in delaying the outgrowth of A431-derived solid tumours. This is the first report describing the successful use of untagged Nanobodies for the in vivo treatment of solid tumours. The results show that functional phage antibody selection, coupled to the rational design of Nanobodies, permits the rapid development of novel anti-cancer antibody-based therapeutics.


Asunto(s)
Anticuerpos/farmacología , Receptores ErbB/antagonistas & inhibidores , Cadenas Pesadas de Inmunoglobulina/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Anticuerpos/aislamiento & purificación , Formación de Anticuerpos , Especificidad de Anticuerpos , Camélidos del Nuevo Mundo/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta Inmunológica , Ensayo de Inmunoadsorción Enzimática , Receptores ErbB/inmunología , Femenino , Humanos , Cadenas Pesadas de Inmunoglobulina/aislamiento & purificación , Ligandos , Ratones , Ratones Desnudos , Sensibilidad y Especificidad , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Biochim Biophys Acta ; 1764(8): 1307-19, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16872921

RESUMEN

With the completion of the sequence of the human genome, emphasis is now switching to the human proteome. However, the number of proteins is not only larger than mRNAs in the transcriptome, proteins need often to be in complex with other proteins to be functional. A favourable option to study proteins in their natural context is with a combination of biochemical and microscopic techniques using specific antibodies. Therefore, we designed a fast, reliable and controllable selection and screening of single-domain antibody fragments (VHH) from a Camelid non-immune library. We isolated VHH for four muscle disease related proteins; emerin, actin, tropomyosin-1, and nuclear poly(A)-binding protein. Important features of antibodies for target validation studies are recognition of the antigen in natural conformations and biologically relevant complexes. We show that selected antibody fragments are functional in various immunological techniques and prove useful in diagnostic applications. Our selection strategy is amenable to automation and to the establishment of proteomics platforms. It opens the way to quickly and cost-effectively obtain multiple antibody fragments for many antigens that can detect changes in their localization, level, and modification as well as subtle changes in supramolecular structures, which often associate with disease.


Asunto(s)
Camélidos del Nuevo Mundo/genética , Camélidos del Nuevo Mundo/inmunología , Fragmentos de Inmunoglobulinas/genética , Actinas/genética , Actinas/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/genética , Afinidad de Anticuerpos , ADN Complementario/genética , Biblioteca de Genes , Humanos , Fragmentos de Inmunoglobulinas/metabolismo , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/metabolismo , Región Variable de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/metabolismo , Técnicas In Vitro , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , Proteína I de Unión a Poli(A)/genética , Proteína I de Unión a Poli(A)/inmunología , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Tropomiosina/genética , Tropomiosina/inmunología
13.
Nat Struct Mol Biol ; 13(1): 85-9, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16327804

RESUMEN

Lactococcus lactis is a Gram-positive bacterium used extensively by the dairy industry for the manufacture of fermented milk products. The double-stranded DNA bacteriophage p2 infects specific L. lactis strains using a receptor-binding protein (RBP) located at the tip of its noncontractile tail. We have solved the crystal structure of phage p2 RBP, a homotrimeric protein composed of three domains: the shoulders, a beta-sandwich attached to the phage; the neck, an interlaced beta-prism; and the receptor-recognition head, a seven-stranded beta-barrel. We used the complex of RBP with a neutralizing llama VHH domain to identify the receptor-binding site. Structural similarity between the recognition-head domain of phage p2 and those of adenoviruses and reoviruses, which invade mammalian cells, suggests that these viruses, despite evolutionary distant targets, lack of sequence similarity and the different chemical nature of their genomes (DNA versus RNA), might have a common ancestral gene.


Asunto(s)
Bacteriófago P2/química , Bacteriófago P2/genética , Lactococcus lactis/virología , Mamíferos/virología , Proteínas Virales/química , Proteínas Virales/genética , Secuencia de Aminoácidos , Animales , Bacteriófago P2/metabolismo , Bacteriófago P2/ultraestructura , Sitios de Unión , Cristalografía por Rayos X , Internet , Microscopía Inmunoelectrónica , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Estructura Cuaternaria de Proteína , Alineación de Secuencia , Proteínas Virales/metabolismo
14.
Hum Mol Genet ; 15(1): 105-11, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16319127

RESUMEN

Oculopharyngeal muscular dystrophy (OPMD) belongs to the group of protein aggregation disorders and is caused by extensions of the N-terminal polyalanine stretch of the nuclear polyA-binding protein 1 (PABPN1). The presence of PABPN1-containing intranuclear aggregates in skeletal muscle is unique for OPMD and is also observed in transgenic mouse and cell models for OPMD. These models consistently support a direct role for the protein aggregation in OPMD pathogenesis. We have isolated and characterized a diverse panel of single-domain antibody reagents (VHH), recognizing different epitopes in PABPN1. The antibody reagents specifically detect endogenous PABPN1 in cell lysates on western blot and label PABPN1 in cultured cells and muscle sections. When expressed intracellularly as intrabodies in a cellular model for OPMD, aggregation of PABPN1 was prevented in a dose-dependent manner. More importantly yet, these intrabodies could also reduce the presence of already existing aggregates. Given the domain specificity of VHH-mediated aggregation interference, this approach at least allows the definition of the nucleation kernel in aggregation-prone proteins, thus facilitating etiological insight into this and other protein aggregation disorders, and ultimately, it may well provide useful therapeutic agents.


Asunto(s)
Anticuerpos/metabolismo , Cuerpos de Inclusión/metabolismo , Músculo Esquelético/patología , Distrofia Muscular Oculofaríngea/tratamiento farmacológico , Proteína II de Unión a Poli(A)/genética , Animales , Anticuerpos/uso terapéutico , Western Blotting , Células COS , Chlorocebus aethiops , Relación Dosis-Respuesta a Droga , Mapeo Epitopo , Células HeLa , Humanos , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/patología
15.
J Bacteriol ; 187(13): 4531-41, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15968064

RESUMEN

Bacteriophage p2 belongs to the most prevalent lactococcal phage group (936) responsible for considerable losses in industrial production of cheese. Immunization of a llama with bacteriophage p2 led to higher titers of neutralizing heavy-chain antibodies (i.e., devoid of light chains) than of the classical type of immunoglobulins. A panel of p2-specific single-domain antibody fragments was obtained using phage display technology, from which a group of potent neutralizing antibodies were identified. The antigen bound by these antibodies was identified as a protein with a molecular mass of 30 kDa, homologous to open reading frame 18 (ORF18) of phage sk1, another 936-like phage for which the complete genomic sequence is available. By the use of immunoelectron microscopy, the protein is located at the tip of the tail of the phage particle. The addition of purified ORF18 protein to a bacterial culture suppressed phage infection. This result and the inhibition of cell lysis by anti-ORF18 protein antibodies support the conclusion that the ORF18 protein plays a crucial role in the interaction of bacteriophage p2 with the surface receptors of Lactococcus lactis.


Asunto(s)
Anticuerpos/farmacología , Proteínas Bacterianas/inmunología , Bacteriófago P2/inmunología , Camélidos del Nuevo Mundo , Lactococcus lactis/virología , Receptores Virales/inmunología , Secuencia de Aminoácidos , Animales , Especificidad de Anticuerpos , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Bacteriólisis/inmunología , Bacteriófago P2/ultraestructura , Tecnología de Alimentos/métodos , Cadenas Pesadas de Inmunoglobulina/farmacología , Lactococcus lactis/química , Microscopía Inmunoelectrónica , Datos de Secuencia Molecular , Peso Molecular , Pruebas de Neutralización , Sistemas de Lectura Abierta , Receptores Virales/genética , Alineación de Secuencia , Proteínas Virales/inmunología , Replicación Viral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...