Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
MAbs ; 11(5): 803-808, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31107637

RESUMEN

Two recent publications out of the same research laboratory report on structure-based in silico design of antibodies against viral targets without sequence disclosure. Cross-referencing the published data to patent databases, we established the sequence identity of said computationally designed antibodies. In both cases, the antibodies align with high sequence identity to previously reported antibodies of the same specificity. This clear underlying sequence relationship, which is far closer than the antibody templates reported to seed the computational design, suggests an alternative origin of the computationally designed antibodies. The lack of both reproducible computational algorithms and of output sequences in the initial publications obscures the relationship to previously reported antibodies, and sows doubt as to the genesis narrative described therein.


Asunto(s)
Anticuerpos Monoclonales/química , Anticuerpos Antivirales/química , Gripe Humana/inmunología , Virus Zika/inmunología , Algoritmos , Secuencia de Aminoácidos , Biología Computacional , Simulación por Computador , Epítopos/inmunología , Humanos
2.
Science ; 351(6277): 1078-83, 2016 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-26912366

RESUMEN

Antibodies targeting the Ebola virus surface glycoprotein (EBOV GP) are implicated in protection against lethal disease, but the characteristics of the human antibody response to EBOV GP remain poorly understood. We isolated and characterized 349 GP-specific monoclonal antibodies (mAbs) from the peripheral B cells of a convalescent donor who survived the 2014 EBOV Zaire outbreak. Remarkably, 77% of the mAbs neutralize live EBOV, and several mAbs exhibit unprecedented potency. Structures of selected mAbs in complex with GP reveal a site of vulnerability located in the GP stalk region proximal to the viral membrane. Neutralizing antibodies targeting this site show potent therapeutic efficacy against lethal EBOV challenge in mice. The results provide a framework for the design of new EBOV vaccine candidates and immunotherapies.


Asunto(s)
Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Neutralizantes/aislamiento & purificación , Anticuerpos Antivirales/aislamiento & purificación , Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/química , Anticuerpos Antivirales/uso terapéutico , Formación de Anticuerpos , Complejo Antígeno-Anticuerpo/química , República Democrática del Congo/epidemiología , Brotes de Enfermedades , Vacunas contra el Virus del Ébola/inmunología , Vacunas contra el Virus del Ébola/uso terapéutico , Fiebre Hemorrágica Ebola/epidemiología , Fiebre Hemorrágica Ebola/terapia , Humanos , Inmunización Pasiva , Ratones , Sobrevivientes , Donantes de Tejidos , Proteínas del Envoltorio Viral/química , Virión/inmunología
3.
Int J Nanomedicine ; 10: 2595-617, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25878495

RESUMEN

Active molecular targeting has become an important aspect of nanoparticle development for oncology indications. Here, we describe molecular targeting of iron oxide nanoparticles (IONPs) to the folate receptor alpha (FOLRα) using an engineered antibody fragment (Ffab). Compared to control nanoparticles targeting the non-relevant botulinum toxin, the Ffab-IONP constructs selectively accumulated on FOLRα-overexpressing cancer cells in vitro, where they exhibited the capacity to internalize into intracellular vesicles. Similarly, Ffab-IONPs homed to FOLRα-positive tumors upon intraperitoneal administration in an orthotopic murine xenograft model of ovarian cancer, whereas negative control particles showed no detectable tumor accumulation. Interestingly, Ffab-IONPs built with custom 120 nm nanoparticles exhibited lower in vitro targeting efficiency when compared to those built with commercially sourced 180 nm nanoparticles. In vivo, however, the two Ffab-IONP platforms achieved equivalent tumor homing, although the smaller 120 nm IONPs were more prone to liver sequestration. Overall, the results show that Ffab-mediated targeting of IONPs yields specific, high-level accumulation within cancer cells, and this fact suggests that Ffab-IONPs could have future utility in ovarian cancer diagnostics and therapy.


Asunto(s)
Anticuerpos , Receptor 1 de Folato , Nanopartículas de Magnetita/química , Neoplasias/metabolismo , Animales , Anticuerpos/química , Anticuerpos/inmunología , Anticuerpos/metabolismo , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Receptor 1 de Folato/inmunología , Receptor 1 de Folato/metabolismo , Humanos , Ratones
4.
PLoS One ; 10(2): e0115636, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25695795

RESUMEN

Realizing the full potential of iron oxide nanoparticles (IONP) for cancer diagnosis and therapy requires selective tumor cell accumulation. Here, we report a systematic analysis of two key determinants for IONP homing to human breast cancers: (i) particle size and (ii) active vs passive targeting. In vitro, molecular targeting to the HER2 receptor was the dominant factor driving cancer cell association. In contrast, size was found to be the key determinant of tumor accumulation in vivo, where molecular targeting increased tumor tissue concentrations for 30 nm but not 100 nm IONP. Similar to the in vitro results, PEGylation did not influence in vivo IONP biodistribution. Thus, the results reported here indicate that the in vitro advantages of molecular targeting may not consistently extend to pre-clinical in vivo settings. These observations may have important implications for the design and clinical translation of advanced, multifunctional, IONP platforms.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Compuestos Férricos/química , Nanopartículas/química , Nanopartículas/uso terapéutico , Animales , Neoplasias de la Mama/genética , Humanos , Ratones , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo
5.
MAbs ; 7(1): 243-54, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25523282

RESUMEN

Staphylococcus aureus is a major human pathogen associated with high mortality. The emergence of antibiotic resistance and the inability of antibiotics to counteract bacterial cytotoxins involved in the pathogenesis of S. aureus call for novel therapeutic approaches, such as passive immunization with monoclonal antibodies (mAbs). The complexity of staphylococcal pathogenesis and past failures with single mAb products represent considerable barriers for antibody-based therapeutics. Over the past few years, efforts have focused on neutralizing α-hemolysin. Recent findings suggest that the concerted actions of several cytotoxins, including the bi-component leukocidins play important roles in staphylococcal pathogenesis. Therefore, we aimed to isolate mAbs that bind to multiple cytolysins by employing high diversity human IgG1 libraries presented on the surface of yeast cells. Here we describe cross-reactive antibodies with picomolar affinity for α-hemolysin and 4 different bi-component leukocidins that share only ∼26% overall amino acid sequence identity. The molecular basis of cross-reactivity is the recognition of a conformational epitope shared by α-hemolysin and F-components of gamma-hemolysin (HlgAB and HlgCB), LukED and LukSF (Panton-Valentine Leukocidin). The amino acids predicted to form the epitope are conserved and known to be important for cytotoxic activity. We found that a single cross-reactive antibody prevented lysis of human phagocytes, epithelial and red blood cells induced by α-hemolysin and leukocidins in vitro, and therefore had superior effectiveness compared to α-hemolysin specific antibodies to protect from the combined cytolytic effect of secreted S. aureus toxins. Such mAb afforded high levels of protection in murine models of pneumonia and sepsis.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales/inmunología , Proteínas Bacterianas/inmunología , Proteínas Hemolisinas/inmunología , Inmunoglobulina G/inmunología , Leucocidinas/inmunología , Staphylococcus aureus/inmunología , Animales , Anticuerpos Antibacterianos/química , Anticuerpos Monoclonales/química , Especificidad de Anticuerpos , Proteínas Bacterianas/química , Línea Celular , Proteínas Hemolisinas/química , Humanos , Inmunoglobulina G/química , Leucocidinas/química , Conejos , Staphylococcus aureus/química
6.
Nat Biotechnol ; 30(8): 742-4, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22871706
7.
Yeast ; 28(3): 237-52, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21360735

RESUMEN

To humanize the glycosylation pathway in the yeast Pichia pastoris, we developed several combinatorial genetic libraries and used them to properly localize active eukaryotic mannosidases and sugar transferases. Here we report the details of the fusion of up to 66 N-terminal targeting sequences of fungal type II membrane proteins to 33 catalytic domains of heterologous glycosylation enzymes. We show that while it is difficult to predict which leader/catalytic domain will result in the desired activity, analysis of the fusion protein libraries allows for the selection of the leader/catalytic domain combinations that function properly. This combinatorial approach, together with a high-throughput screening protocol, has allowed us to humanize the yeast glycosylation pathway to secrete human glycoprotein with complex N-glycosylation.


Asunto(s)
Retículo Endoplásmico/enzimología , Glucosiltransferasas/metabolismo , Aparato de Golgi/enzimología , Manosidasas/metabolismo , Pichia/enzimología , Ingeniería de Proteínas , Glucosiltransferasas/genética , Manosidasas/genética , Pichia/genética , Señales de Clasificación de Proteína/genética , Transporte de Proteínas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
8.
Bioconjug Chem ; 22(5): 903-12, 2011 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-21395336

RESUMEN

The site-specific modification of proteins is expected to be an important capability for the synthesis of bioconjugates in the future. However, the traditional repertoire of reactions available for the direct modification of proteins suffers from lack of specificity, necessitating costly downstream processing to isolate the specific species of interest. (1) Here, we use a well-established, glycan-specific chemistry to PEGylate model glycoproteins, each containing a unique reactive GalNAc attached to a specifically engineered threonine residue. By engineering E. coli to execute the initial steps of human, mucin-type O-glycosylation, we were able to obtain homogeneous site-specifically modified glycoproteins with fully human glycan linkages. Two mucin-based reporters as well as several fusion proteins containing eight-amino-acid GalNAc-T recognition sequences were glycosylated in this engineered glycocompetent strain of E. coli. The use of one sequence in particular, PPPTSGPT, resulted in site-specific glycan occupancy of approximately 69% at the engineered threonine. The GalNAc present on the purified glycoprotein was oxidized by galactose oxidase and then coupled to hydroxylamine functionalized 20 kDa PEG in the presence of aniline. The glycoprotein could be converted to the PEGylated product at approximately 85% yield and >98% purity as determined by comparison to the products of control reactions.


Asunto(s)
Escherichia coli/metabolismo , Glicoproteínas/biosíntesis , Glicoproteínas/química , Ingeniería de Proteínas/métodos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Conformación de Carbohidratos , Galactosa Oxidasa/química , Galactosa Oxidasa/metabolismo , Glicosilación , Humanos , Oxidación-Reducción , Polietilenglicoles/química , Polietilenglicoles/metabolismo , Polisacáridos/química , Polisacáridos/metabolismo , Treonina/química , Treonina/metabolismo
9.
Methods Mol Biol ; 534: 213-23, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19277549

RESUMEN

The production of recombinant therapeutic glycoproteins is an active area of research and drug development. Typically, improvements in therapeutic glycoprotein efficacy have focused on engineering additional N-glycosylation sites into the primary amino acid sequence or attempting to control a particular glycoform profile on a protein through process improvements. Recently, a number of alternative expression systems have appeared that are challenging the dominance of mammalian cell culture. Our laboratory has focused on the re-engineering of the secretory pathway in the yeast Pichia pastoris to perform glycosylation reactions that mimic processing of N-glycans in humans. We have demonstrated that human antibodies with specific human N-glycan structures can be produced in glycoengineered lines of Pichia pastoris and that antibody-mediated effector functions can be optimized by generating specific glycoforms. In this chapter we provide detailed protocols for the analysis of glycosylation on intact glycoproteins by MALDI-TOF and site specific N-glycan occupancy on digested glycoprotein using ESI-MS.


Asunto(s)
Glicoproteínas/metabolismo , Glicosilación , Pichia/metabolismo , Proteínas Recombinantes/metabolismo , Espectrometría de Masa por Ionización de Electrospray/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Animales , Cromatografía Líquida de Alta Presión/métodos , Glicoproteínas/química , Humanos , Proteínas Recombinantes/química
10.
Nat Protoc ; 3(6): 1026-31, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18546597

RESUMEN

O-glycosylation is a post-translational protein modification that occurs in all eukaryotes. Yeasts have received increasing attention as a host for therapeutic protein production because of their ability to secrete high levels of recombinant protein. Because yeasts such as Pichia pastoris have been shown to O-glycosylate some proteins with varying effects on protein function, it is important to elucidate the nature of this modification. Methods that characterize O-glycosylation on a qualitative and quantitative basis are thus important when considering yeast as a host for therapeutic protein production. This protocol describes the release of O-glycans from a protein sample by -elimination under alkaline conditions using sodium borohydride and sodium hydroxide. The released O-linked oligosaccharides are subsequently processed and then separated by high-performance anion exchange chromatography with pulsed amperometric detection (HPAEC-PAD). An estimation of O-glycan molar occupancy and average O-mannose chain length is ultimately derived. This protocol requires approximately 3 d for completion. This method provides an assessment of O-glycosylation and allows one to correlate the effect of O-glycosylation on protein properties.


Asunto(s)
Cromatografía por Intercambio Iónico/métodos , Micología/métodos , Polisacáridos/análisis , Levaduras/química , Cromatografía Líquida de Alta Presión/métodos , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Glicoproteínas/química , Glicoproteínas/metabolismo , Glicosilación , Monosacáridos/análisis , Polisacáridos/metabolismo , Procesamiento Proteico-Postraduccional , Levaduras/metabolismo
11.
Methods Mol Biol ; 389: 99-106, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17951637

RESUMEN

With an ever increasing number of proteins being expressed in the Pichia system, there is a growing need to rapidly develop scalable and robust purification schemes. This chapter describes a high-throughput method to screen for the optimal chromatography conditions and resin to capture and release a protein secreted by Pichia pastoris. The method involves a chromatography matrix involving four resins (Q-Sepharose, DEAE-Sepharose, SP-Sepharose, and CMSepharose), 4 pHs from 5.0 to 8.0, and 3 NaCl concentrations. The method was tested on three proteins and found to be reproducible and easily scalable.


Asunto(s)
Proteínas Fúngicas/aislamiento & purificación , Pichia/metabolismo , Resinas Sintéticas/metabolismo , Cromatografía por Intercambio Iónico , Ensayo de Inmunoadsorción Enzimática
12.
Methods Mol Biol ; 389: 139-50, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17951640

RESUMEN

Our laboratory has focused on the re-engineered of the secretory pathway of Pichia pastoris to perform glycosylation reactions that mimic processing of N-glycans in humans and other higher mammals (1,2). A reporter protein with a single N-linked glycosylation site, a His-tagged Kringle 3 domain of human plasminogen (K3), was used to identify combinations of optimal leader/catalytic domain(s) to recreate human N-glycan processing in the Pichia system. In this chapter we describe detailed protocols for high-throughput purification of K3, enzymatic release of N-glycans, matrix-assisted laser desorption ionization time-of-flight and high-performance liquid chromatography analysis of the released N-glycans. The developed protocols can be adapted to the characterization of N-glycans from any purified protein expressed in P. pastoris.


Asunto(s)
Polisacáridos/análisis , Polisacáridos/química , Proteínas/química , Cromatografía de Afinidad , Cromatografía Líquida de Alta Presión , Glicósido Hidrolasas/metabolismo , Humanos , Kringles , Oligosacáridos/análisis , Pichia , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
13.
Curr Opin Biotechnol ; 18(5): 387-92, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17951046

RESUMEN

Yeasts have been extensively used as model organisms to elucidate cellular processes and their mechanism in lower eukaryotes. Consequently, a large number of powerful genetic tools have been developed to engineer yeast and improve its utility. These tools and the development of efficient large-scale fermentation processes have made recombinant protein expression in yeast an attractive choice. However, for the production of glycoproteins for human use, native high-mannose yeast glycosylation is not suitable and therefore represents a major limitation for yeast based protein expression systems. Over the last two decades several groups have attempted to overcome this problem, yet with limited success. Recently however, major advances in the glycoengineering of the yeast Pichia pastoris, have culminated in the production of fully humanized sialylated glycoproteins.


Asunto(s)
Glicoproteínas/metabolismo , Pichia/fisiología , Ingeniería de Proteínas/tendencias , Proteínas Recombinantes/metabolismo , Transfección/métodos , Animales , Mejoramiento Genético/métodos , Glicoproteínas/genética , Glicosilación , Humanos
14.
Science ; 313(5792): 1441-3, 2006 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-16960007

RESUMEN

Yeast is a widely used recombinant protein expression system. We expanded its utility by engineering the yeast Pichia pastoris to secrete human glycoproteins with fully complex terminally sialylated N-glycans. After the knockout of four genes to eliminate yeast-specific glycosylation, we introduced 14 heterologous genes, allowing us to replicate the sequential steps of human glycosylation. The reported cell lines produce complex glycoproteins with greater than 90% terminal sialylation. Finally, to demonstrate the utility of these yeast strains, functional recombinant erythropoietin was produced.


Asunto(s)
Eritropoyetina/metabolismo , Pichia/genética , Ingeniería de Proteínas , Sialoglicoproteínas/biosíntesis , Animales , Línea Celular , Clonación Molecular , Ácido N-Acetilneuramínico Citidina Monofosfato/metabolismo , Eritropoyetina/química , Eritropoyetina/genética , Vectores Genéticos , Glicosilación , Humanos , Pichia/metabolismo , Ratas , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Ácidos Siálicos/metabolismo , Sialoglicoproteínas/química , Sialoglicoproteínas/genética , Transformación Genética
15.
Nat Biotechnol ; 24(2): 210-5, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16429149

RESUMEN

As the fastest growing class of therapeutic proteins, monoclonal antibodies (mAbs) represent a major potential drug class. Human antibodies are glycosylated in their native state and all clinically approved mAbs are produced by mammalian cell lines, which secrete mAbs with glycosylation structures that are similar, but not identical, to their human counterparts. Glycosylation of mAbs influences their interaction with immune effector cells that kill antibody-targeted cells. Here we demonstrate that human antibodies with specific human N-glycan structures can be produced in glycoengineered lines of the yeast Pichia pastoris and that antibody-mediated effector functions can be optimized by generating specific glycoforms. Glycoengineered P. pastoris provides a general platform for producing recombinant antibodies with human N-glycosylation.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Mejoramiento Genético/métodos , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/genética , Pichia/genética , Pichia/metabolismo , Ingeniería de Proteínas/métodos , Anticuerpos Monoclonales/genética , Glicosilación , Humanos , Proteínas Recombinantes/biosíntesis
16.
Protein Expr Purif ; 46(2): 179-88, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16249097

RESUMEN

Self-assembling peptides have emerged as an attractive scaffold material for tissue engineering, yet the expense associated with solid phase chemical synthesis has limited their broad use. In addition, the fidelity of chemical synthesis constrains the length of polypeptides that can be produced homogeneously by this method. Template-derived biosynthesis by recombinant DNA technology may overcome both of these problems. However, recovery of polypeptides from recombinant protein expression systems typically involves multi-step purification schemes. In this study, we report an integrated approach to recombinantly produce and purify self-assembling peptides from the recently developed expression host Ralstonia eutropha. The purification is based on the specific affinity of carbohydrate binding modules (CBMs) to cellulose. In a first step, we identified CBMs that express well in R. eutropha by assembling a fusion library of green fluorescent protein (GFP) and CBMs and determining the fluorescence of cell-free extracts. Three GFP::CBM fusions were found to express at levels similar to GFP alone, of which two CBMs were able to mediate cellulose binding of the GFP::CBM fusion. These two CBMs were then fused to multiple repeats of the self-assembling peptide RAD16-I::E (N-RADARADARADARADAE-C). The fusion protein CBM::E::(RAD16-I::E)4 was expressed in R. eutropha and purified using the CBM's affinity for cellulose. Subsequent proteolytic cleavage with endoproteinase GluC liberated RAD16-I::E peptide monomers with similar properties to the chemically synthesized counterpart RAD16-I.


Asunto(s)
Proteínas Portadoras/biosíntesis , Cupriavidus necator/genética , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Portadoras/genética , Proteínas Portadoras/aislamiento & purificación , Cupriavidus necator/crecimiento & desarrollo , Expresión Génica , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Ingeniería de Tejidos/métodos
17.
Appl Environ Microbiol ; 71(10): 5735-42, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16204482

RESUMEN

Protein purification of recombinant proteins constitutes a significant cost of biomanufacturing and various efforts have been directed at developing more efficient purification methods. We describe a protein purification scheme wherein Ralstonia eutropha is used to produce its own "affinity matrix," thereby eliminating the need for external chromatographic purification steps. This approach is based on the specific interaction of phasin proteins with granules of the intracellular polymer polyhydroxybutyrate (PHB). By creating in-frame fusions of phasins and green fluorescent protein (GFP) as a model protein, we demonstrated that GFP can be efficiently sequestered to the surface of PHB granules. In a second step, we generated a phasin-intein-GFP fusion, wherein the self-cleaving intein can be activated by the addition of thiols. This construct allowed for the controlled binding and release of essentially pure GFP in a single separation step. Finally, pure, active beta-galactosidase was obtained in a single step using the above described method.


Asunto(s)
Cupriavidus necator/metabolismo , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biotecnología/métodos , Cupriavidus necator/genética , Cupriavidus necator/crecimiento & desarrollo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ditiotreitol , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hidroxibutiratos/metabolismo , Inteínas/genética , Poliésteres/metabolismo , Proteínas Recombinantes de Fusión/genética
18.
Protein Sci ; 14(6): 1387-95, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15883185

RESUMEN

This work combines two well-established technologies to generate a breakthrough in protein production and purification. The first is the production of polyhydroxybutyrate (PHB) granules in engineered strains of Escherichia coli. The second is a recently developed group of self-cleaving affinity tags based on protein splicing elements known as inteins. By combining these technologies with a PHB-specific binding protein, a self-contained protein expression and purification system has been developed. In this system, the PHB-binding protein effectively acts as an affinity tag for desired product proteins. The tagged product proteins are expressed in E. coli strains that also produce intracellular PHB granules, where they bind to the granules via the PHB-binding tag. The granules and attached proteins can then be easily recovered following cell lysis by simple mechanical means. Once purified, the product protein is self-cleaved from the granules and released into solution in a substantially purified form. This system has been successfully used at laboratory scale to purify several active test proteins at reasonable yield. By allowing the bacterial cells to effectively produce both the affinity resin and tagged target protein, the cost associated with the purification of recombinant proteins could be greatly reduced. It is expected that this combination of improved economics and simplicity will constitute a significant breakthrough in both large-scale production of purified proteins and enzymes and high-throughput proteomics studies of peptide libraries.


Asunto(s)
Proteínas Bacterianas/química , Gránulos Citoplasmáticos/química , Proteínas de Unión al ADN/química , Escherichia coli/química , Inteínas , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Bacterianas/genética , Cromatografía de Afinidad/métodos , Clonación Molecular , Gránulos Citoplasmáticos/genética , Gránulos Citoplasmáticos/ultraestructura , Proteínas de Unión al ADN/genética , Escherichia coli/genética , Escherichia coli/ultraestructura , Expresión Génica , Vectores Genéticos , Hidroxibutiratos/química , Inteínas/genética , Proteínas Recombinantes de Fusión/química
19.
Glycobiology ; 15(6): 615-24, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15677381

RESUMEN

Rat endomannosidase is a glycosidic enzyme that catalyzes the cleavage of di-, tri-, or tetrasaccharides (Glc(1-3)Man), from N-glycosylation intermediates with terminal glucose residues. To date it is the only characterized member of this class of endomannosidic enzymes. Although this protein has been demonstrated to localize to the Golgi lumenal membrane, the mechanism by which this occurs has not yet been determined. Using the rat endomannosidase sequence, we identified three homologs, one each in the human, mouse, and rat genomes. Alignment of the four encoded protein sequences demonstrated that the newly identified sequences are highly conserved but differed significantly at the N-terminus from the previously reported protein. In this study we have cloned two novel endomannosidase sequences from rat and human cDNA libraries, but were unable to amplify the open reading frame of the previously reported rat sequence. Analysis of the rat genome confirmed that the 59- and 39-termini of the previously reported sequence were in fact located on different chromosomes. This, in combination with our inability to amplify the previously reported sequence, indicated that the N-terminus of the rat endomannosidase sequence previously published was likely in error (a cloning artifact), and that the sequences reported in the current study encode the intact proteins. Furthermore, unlike the previous sequence, the three ORFs identified in this study encode proteins containing a single N-terminal transmembrane domain. Here we demonstrate that this region is responsible for Golgi localization and in doing so confirm that endomannosidase is a type II membrane protein, like the majority of other secretory pathway glycosylation enzymes.


Asunto(s)
Manosidasas/genética , Proteínas de la Membrana/genética , Secuencia de Aminoácidos , Animales , Clonación Molecular , Regulación Enzimológica de la Expresión Génica , Vectores Genéticos/genética , Humanos , Manosidasas/biosíntesis , Manosidasas/clasificación , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/clasificación , Ratones , Datos de Secuencia Molecular , Pichia/genética , Ratas , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...