Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Annu Rev Immunol ; 36: 695-715, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29490163

RESUMEN

The unique class of heavy chain-only antibodies, present in Camelidae, can be shrunk to just the variable region of the heavy chain to yield VHHs, also called nanobodies. About one-tenth the size of their full-size counterparts, nanobodies can serve in applications similar to those for conventional antibodies, but they come with a number of signature advantages that find increasing application in biology. They not only function as crystallization chaperones but also can be expressed inside cells as such, or fused to other proteins to perturb the function of their targets, for example, by enforcing their localization or degradation. Their small size also affords advantages when applied in vivo, for example, in imaging applications. Here we review such applications, with particular emphasis on those areas where conventional antibodies would face a more challenging environment.


Asunto(s)
Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/inmunología , Animales , Formación de Anticuerpos , Técnicas de Visualización de Superficie Celular , Ingeniería Genética , Humanos , Cadenas Pesadas de Inmunoglobulina/biosíntesis , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/inmunología , Anticuerpos de Dominio Único/biosíntesis , Anticuerpos de Dominio Único/uso terapéutico , Relación Estructura-Actividad
2.
J Biol Chem ; 299(9): 104927, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37330175

RESUMEN

Methicillin-resistant Staphylococcus aureus, or MRSA, is one of the major causative agents of hospital-acquired infections worldwide. Novel antimicrobial strategies efficient against antibiotic-resistant strains are necessary and not only against S. aureus. Among those, strategies that aim at blocking or dismantling proteins involved in the acquisition of essential nutrients, helping the bacteria to colonize the host, are intensively studied. A major route for S. aureus to acquire iron from the host organism is the Isd (iron surface determinant) system. In particular, the hemoglobin receptors IsdH and IsdB located on the surface of the bacterium are necessary to acquire the heme moiety containing iron, making them a plausible antibacterial target. Herein, we obtained an antibody of camelid origin that blocked heme acquisition. We determined that the antibody recognized the heme-binding pocket of both IsdH and IsdB with nanomolar order affinity through its second and third complementary-determining regions. The mechanism explaining the inhibition of acquisition of heme in vitro could be described as a competitive process in which the complementary-determining region 3 from the antibody blocked the acquisition of heme by the bacterial receptor. Moreover, this antibody markedly reduced the growth of three different pathogenic strains of MRSA. Collectively, our results highlight a mechanism for inhibiting nutrient uptake as an antibacterial strategy against MRSA.


Asunto(s)
Anticuerpos Antibacterianos , Staphylococcus aureus Resistente a Meticilina , Receptores de Superficie Celular , Anticuerpos de Dominio Único , Humanos , Antibacterianos/farmacología , Hemo/metabolismo , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Receptores de Superficie Celular/metabolismo , Receptores de Superficie Celular/uso terapéutico , Anticuerpos de Dominio Único/biosíntesis , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/metabolismo , Anticuerpos de Dominio Único/farmacología , Infecciones Estafilocócicas/tratamiento farmacológico , Antígenos Bacterianos/inmunología , Anticuerpos Antibacterianos/genética , Anticuerpos Antibacterianos/inmunología , Camélidos del Nuevo Mundo , Animales , Unión Proteica/efectos de los fármacos , Modelos Moleculares , Simulación de Dinámica Molecular
3.
Protein Expr Purif ; 222: 106522, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38851552

RESUMEN

OBJECTIVE: To screen and obtain specific anti-lymphocyte activation gene-3 (LAG3) nanobody sequences, purify and express recombinant anti-LAG3 nanobody, and verify its effect on promoting T cells to kill tumor cells. METHODS: Based on the camel derived natural nanobody phage display library constructed by the research group, the biotinylated LAG3 antigen was used as the target, and the anti-LAG3 nanobody sequences were screened by biotin-streptavidin liquid phase screening, phage-ELISA and sequencing. The sequence-conjµgated human IgG1 Fc fragment was obtained, the recombinant anti-LAG3 nanobody expression vector was constructed, the expression of the recombinant anti-LAG3 nanobody was induced by IPTG and purified, and the characteristics and functions of the recombinant anti-LAG3 nanobody were verified by SDS-PAGE, Western blot, cytotoxicity assay, etc. RESULTS: One anti-LAG3 nanobody sequence was successfully screened, and the corresponding recombinant anti-LAG3 nanobody-expressing bacteria were constructed. The results of SDS-PAGE, Western blot and cytotoxicity assay showed that the recombinant anti-LAG3 nanobody was successfully expressed, which was specific, and it could promote the killing ability of T cells against tumor cells, and the optimal concentration was 200 µg/mL. CONCLUSION: The recombinant anti-LAG3 nanobody screened and expressed has specific and auxiliary anti-tumor cell effects, which lays a foundation for its subsequent application.


Asunto(s)
Proteína del Gen 3 de Activación de Linfocitos , Anticuerpos de Dominio Único , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/biosíntesis , Anticuerpos de Dominio Único/farmacología , Humanos , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos CD/metabolismo , Antígenos CD/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/química , Animales , Biblioteca de Péptidos , Camelus/inmunología , Camelus/genética , Línea Celular Tumoral , Escherichia coli/genética , Linfocitos T/inmunología , Expresión Génica
4.
Protein Expr Purif ; 221: 106501, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38782081

RESUMEN

Nanobodies are single-variable domain antibodies with excellent properties, which are evolving as versatile tools to guide cognate antigens in vitro and in vivo for biological research, diagnosis, and treatment. Given their simple structure, nanobodies are readily produced in multiple systems. However, selecting an appropriate expression system is crucial because different conditions might cause proteins to produce different folds or post-translational modifications (PTMs), and these differences often result in different functions. At present, the strategies of PTMs are rarely reported. The GFP nanobody can specifically target the GFP protein. Here, we engineered a GFP nanobody fused with 6 × His tag and Fc tag, respectively, and expressed in bacteria and mammalian cells. The 6 × His-GFP-nanobody was produced from Escherichia coli at high yields and the pull-down assay indicated that it can precipitate the GFP protein. Meanwhile, the Fc-GFP-nanobody can be expressed in HEK293T cells, and the co-immunoprecipitation experiment can trace and target the GFP-tagged protein in vivo. Furthermore, some different PTMs in antigen-binding regions have been identified after using mass spectrometry (MS) to analyze the GFP nanobodies, which are expressed in prokaryotes and eukaryotes. In this study, a GFP nanobody was designed, and its binding ability was verified by using the eukaryotic and prokaryotic protein expression systems. In addition, this GFP nanobody was transformed into a useful instrument for more in-depth functional investigations of GFP fusion proteins. MS was further used to explore the reason for the difference in binding ability, providing a novel perspective for the study of GFP nanobodies and protein expression purification.


Asunto(s)
Escherichia coli , Proteínas Fluorescentes Verdes , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes de Fusión , Anticuerpos de Dominio Único , Humanos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Fluorescentes Verdes/química , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/biosíntesis , Anticuerpos de Dominio Único/aislamiento & purificación , Anticuerpos de Dominio Único/inmunología , Células HEK293 , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Ingeniería de Proteínas/métodos , Expresión Génica
5.
FASEB J ; 36(4): e22222, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35218573

RESUMEN

Cellular uptake of vitamin B12 in humans is mediated by the endocytosis of the B12 carrier protein transcobalamin (TC) via its cognate cell surface receptor TCblR, encoded by the CD320 gene. Because CD320 expression is associated with the cell cycle and upregulated in highly proliferating cells including cancer cells, this uptake route is a potential target for cancer therapy. We developed and characterized four camelid nanobodies that bind holo-TC (TC in complex with B12 ) or the interface of the human holo-TC:TCblR complex with nanomolar affinities. We determined X-ray crystal structures of these nanobodies bound to holo-TC:TCblR, which enabled us to map their binding epitopes. When conjugated to the model toxin saporin, three of our nanobodies caused growth inhibition of HEK293T cells and therefore have the potential to inhibit the growth of human cancer cells. We visualized the cellular binding and endocytic uptake of the most potent nanobody (TC-Nb4) using fluorescent light microscopy. The co-crystal structure of holo-TC:TCblR with another nanobody (TC-Nb34) revealed novel features of the interface of TC and the LDLR-A1 domain of TCblR, rationalizing the decrease in the affinity of TC-B12 binding caused by the Δ88 mutation in CD320.


Asunto(s)
Anticuerpos Monoclonales/química , Inmunoconjugados/farmacología , Receptores de Superficie Celular/metabolismo , Saporinas/química , Anticuerpos de Dominio Único/química , Transcobalaminas/metabolismo , Vitamina B 12/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Camélidos del Nuevo Mundo , Ciclo Celular , Proliferación Celular , Células HEK293 , Humanos , Inmunoconjugados/química , Inmunoconjugados/inmunología , Inmunotoxinas/química , Inmunotoxinas/farmacología , Receptores de Superficie Celular/química , Receptores de Superficie Celular/genética , Saporinas/inmunología , Anticuerpos de Dominio Único/biosíntesis , Anticuerpos de Dominio Único/inmunología
6.
Acta Pharmacol Sin ; 44(5): 1095-1104, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36418428

RESUMEN

CKLF (chemokine-like factor)-MARVEL transmembrane domain containing protein 6 (CMTM6) is a novel regulator to maintain the stability of PD-L1. CMTM6 can colocalize and interact with PD-L1 on the recycling endosomes and cell membrane, preventing PD-L1 from lysosome-mediated degradation and proteasome-mediated degradation thus increasing the half-life of PD-L1 on the cell membrane. The difficulties in obtaining stable full-length PD-L1 and CMTM6 proteins hinder the research on their structures, function as well as related drug development. Using lauryl maltose neopentyl glycol (LMNG) as the optimized detergent and a cell membrane mimetic strategy, we assembled a stable membrane-bound full-length CMTM6-PD-L1 complex with amphipol A8-35. When the PD-1/PD-L1-CMTM6 interactions were analyzed, we found that CMTM6 greatly enhanced the binding and delayed the dissociation of PD-1/PD-L1, thus affecting immunosuppressive signaling and anti-apoptotic signaling. We then used the CMTM6-PD-L1 complex as immunogens to generate immune repertoires in camels, and identified a functional anti-CMTM6 nanobody, called 1A5. We demonstrated that the anti-CMTM6 nanobody greatly decreased T-cell immunosuppression and promoted apoptotic susceptibility of tumor cells in vitro, and mainly relied on the cytotoxic effect of CD8+ T-cells to exert tumor growth inhibitory effects in CT26 tumor-bearing mice. In conclusion, the stable membrane-bound full-length CMTM6-PD-L1 complex has been successfully used in studying PD-1/PD-L1-CMTM6 interactions and CMTM6-targeting drug development, suggesting CMTM6 as a novel tumor immunotherapy target.


Asunto(s)
Antígeno B7-H1 , Proteínas con Dominio MARVEL , Neoplasias , Anticuerpos de Dominio Único , Animales , Ratones , Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/metabolismo , Neoplasias/inmunología , Neoplasias/terapia , Receptor de Muerte Celular Programada 1/metabolismo , Proteínas con Dominio MARVEL/inmunología , Proteínas con Dominio MARVEL/metabolismo , Ingeniería de Proteínas/métodos , Anticuerpos de Dominio Único/biosíntesis
7.
Biochem J ; 478(3): 579-595, 2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33480416

RESUMEN

Surface-associated proteins play critical roles in the Plasmodium parasite life cycle and are major targets for vaccine development. The 6-cysteine (6-cys) protein family is expressed in a stage-specific manner throughout Plasmodium falciparum life cycle and characterized by the presence of 6-cys domains, which are ß-sandwich domains with conserved sets of disulfide bonds. Although several 6-cys family members have been implicated to play a role in sexual stages, mosquito transmission, evasion of the host immune response and host cell invasion, the precise function of many family members is still unknown and structural information is only available for four 6-cys proteins. Here, we present to the best of our knowledge, the first crystal structure of the 6-cys protein Pf12p determined at 2.8 Šresolution. The monomeric molecule folds into two domains, D1 and D2, both of which adopt the canonical 6-cys domain fold. Although the structural fold is similar to that of Pf12, its paralog in P. falciparum, we show that Pf12p does not complex with Pf41, which is a known interaction partner of Pf12. We generated 10 distinct Pf12p-specific nanobodies which map into two separate epitope groups; one group which binds within the D2 domain, while several members of the second group bind at the interface of the D1 and D2 domain of Pf12p. Characterization of the structural features of the 6-cys family and their associated nanobodies provide a framework for generating new tools to study the diverse functions of the 6-cys protein family in the Plasmodium life cycle.


Asunto(s)
Antígenos de Protozoos/química , Anticuerpos de Dominio Único/inmunología , Secuencia de Aminoácidos , Animales , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Antígenos de Protozoos/metabolismo , Sitios de Unión , Western Blotting , Camélidos del Nuevo Mundo/inmunología , Cristalografía por Rayos X , Ensayo de Inmunoadsorción Enzimática , Epítopos/inmunología , Interferometría , Modelos Moleculares , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Plasmodium falciparum/metabolismo , Conformación Proteica , Dominios Proteicos , Mapeo de Interacción de Proteínas , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Anticuerpos de Dominio Único/biosíntesis , Anticuerpos de Dominio Único/aislamiento & purificación
8.
Biochem Biophys Res Commun ; 549: 105-112, 2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33667707

RESUMEN

Escherichia coli is one of the most popularly used hosts to produce recombinant proteins. Most recombinant proteins are produced in the cytoplasm and periplasm, requiring multiple steps to extract and purify recombinant proteins. The Serratia marcescens Lip system (LipB-LipC-LipD) is a type 1 secretion system that selectively secretes LipA from the intracellular to extracellular space in a single step. This study aimed to establish a secretory production system for nanobodies, camelid-derived small molecule antibody fragments, using the S. marcescens Lip system. Surprisingly, E. coli harboring only LipC, a membrane fusion protein of the Lip system, could secrete an anti-green fluorescent protein (GFP)-Nb, a nanobody against GFP, without the addition of a long amino acid sequence. The LipC-based secretion system recognized the Val-Thr-Val sequence at the C-terminus of the nanobody. Finally, Strep-tagged anti-GFP-Nb was purified from culture supernatants of E. coli harboring LipC by Strep-affinity chromatography at a final yield of >5 mg per liter of culture supernatant. These results potently supported that the S. marcescens LipC-based secretion system has the potential to establish an efficient secretory production system for nanobodies.


Asunto(s)
Proteínas Bacterianas/metabolismo , Escherichia coli/metabolismo , Serratia marcescens/metabolismo , Anticuerpos de Dominio Único/biosíntesis , Secuencia de Aminoácidos , Animales , Antígenos/metabolismo , Camelus , Medios de Cultivo , Proteínas Fluorescentes Verdes/metabolismo , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/aislamiento & purificación
9.
Biochem Biophys Res Commun ; 581: 38-45, 2021 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-34653677

RESUMEN

A detrimental role of the receptor for the advanced glycation end product (RAGE) has been identified in the immune response, and various pathological conditions and its V and C1 domains in the extracellular region of RAGE are believed to be the main ligand-binding domains. Consequently, specific inhibitors targeting those domains could be of clinical value in fighting against the pathological condition associated with RAGE over-activation. Single-domain antibodies, also called nanobodies (Nbs), are antibody fragments engineered from the heavy-chain only antibodies found in camelids, which offer a range of advantages in therapy. In this study, we report the development and characterization of the V-C1 domain-specific Nbs. Three Nbs (3CNB, 4BNB, and 5ENB) targeting V-C1 domain of human RAGE were isolated from an immunized alpaca using a phage display. All of these Nbs revealed high thermostability. 3CNB, 4BNB, and 5ENB bind to V-C1 domain with a dissociation constant (KD) of 27.25, 39.37, and 47.85 nM, respectively, using Isothermal Titration Calorimetry (ITC). After homodimerization using human IgG1-Fc fusion, their binding affinity improved to 0.55, 0.62, and 0.41 nM, respectively, using Surface Plasmon Resonance (SPR). Flow cytometry showed all the Fc fusions Nbs can bind to human RAGE expressed on the cell surface. Competitive ELISA further confirmed their V-C1-hS100B blocking ability in solution, providing insights into the applicability of Nbs in treating RAGE-associated diseases.


Asunto(s)
Productos Finales de Glicación Avanzada/química , Fragmentos Fc de Inmunoglobulinas/química , Inmunoglobulina G/química , Receptor para Productos Finales de Glicación Avanzada/química , Proteínas Recombinantes de Fusión/química , Anticuerpos de Dominio Único/biosíntesis , Secuencia de Aminoácidos , Animales , Sitios de Unión , Camélidos del Nuevo Mundo , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Productos Finales de Glicación Avanzada/genética , Productos Finales de Glicación Avanzada/inmunología , Células HEK293 , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Biblioteca de Péptidos , Unión Proteica , Dominios Proteicos , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Receptor para Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/aislamiento & purificación
10.
PLoS Biol ; 16(4): e2005473, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29621251

RESUMEN

We describe the development and application of a suite of modular tools for high-resolution detection of proteins and intracellular protein complexes by electron microscopy (EM). Conditionally stable GFP- and mCherry-binding nanobodies (termed csGBP and csChBP, respectively) are characterized using a cell-free expression and analysis system and subsequently fused to an ascorbate peroxidase (APEX) enzyme. Expression of these cassettes alongside fluorescently labelled proteins results in recruitment and stabilisation of APEX, whereas unbound APEX nanobodies are efficiently degraded by the proteasome. This greatly simplifies correlative analyses, enables detection of less-abundant proteins, and eliminates the need to balance expression levels between fluorescently labelled and APEX nanobody proteins. Furthermore, we demonstrate the application of this system to bimolecular complementation ('EM split-fluorescent protein'), for localisation of protein-protein interactions at the ultrastructural level.


Asunto(s)
Ascorbato Peroxidasas/genética , Células Epiteliales/ultraestructura , Microscopía Electrónica/métodos , Microscopía Fluorescente/métodos , Imagen Individual de Molécula/métodos , Anticuerpos de Dominio Único/química , Animales , Ascorbato Peroxidasas/metabolismo , Línea Celular , Sistema Libre de Células , Cricetulus , Células Epiteliales/metabolismo , Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas , Estabilidad Proteica , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Anticuerpos de Dominio Único/biosíntesis , Anticuerpos de Dominio Único/genética , Proteína Fluorescente Roja
11.
Protein Expr Purif ; 185: 105906, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33991675

RESUMEN

Nanobodies are single-domain antibody constructs derived from the variable regions of heavy chain only (VHH) camelid IgGs. Their small size and single gene format make them amenable to various molecular biology applications that require a protein affinity-based approach. These features, in addition to their high solubility, allows their periplasmic expression, extraction and purification in E. coli systems with relative ease, using standardized protocols. However, some Nanobodies are recalcitrant to periplasmic expression, extraction and purification within E. coli systems. To improve their expression would require either a change in the expression host, vector or an increased scale of expression, all of which entail an increase in the complexity of their expression, and production cost. However, as shown here, specific changes in the existing standard E. coli culture protocol, aimed at reducing breakdown of selective antibiotic pressure, increasing the initial culture inoculum and improving transport to the periplasmic space, rescued the expression of several such refractory Nanobodies. The periplasmic extraction protocol was also changed to ensure efficient osmolysis, prevent both protein degradation and prevent downstream chelation of Ni2+ ions during IMAC purification. Adoption of this protocol will lead to an improvement of the expression of Nanobodies in general, and specifically, those that are recalcitrant.


Asunto(s)
Escherichia coli/metabolismo , Periplasma/metabolismo , Proteínas Recombinantes/aislamiento & purificación , Anticuerpos de Dominio Único/biosíntesis , Secuencia de Aminoácidos , Clonación Molecular , Medios de Cultivo/química , Medios de Cultivo/farmacología , Escherichia coli/genética , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Presión Osmótica , Periplasma/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/genética , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/aislamiento & purificación
12.
Protein Expr Purif ; 184: 105888, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33857600

RESUMEN

Anti-EGFR nanobodies have been successfully applied as antitumor moieties in the photodynamic therapy and drug delivery systems. But the yields of nanobodies were still limited due to the volumetric capacity of the periplasmic compartments and inclusion bodies of Escherichia coli. A comparative study of Pichia pastoris and Escherichia coli was done through characterizing their products. Nanobody 7D12 and 7D12-9G8 were successfully expressed in Pichia pastoris with 6-13.6-fold higher yield. Both two types of nanobodies had internalization ability to be developed as antitumor moieties.


Asunto(s)
Antineoplásicos Inmunológicos , Escherichia coli , Proteínas de Neoplasias , Saccharomycetales , Anticuerpos de Dominio Único , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/aislamiento & purificación , Antineoplásicos Inmunológicos/farmacología , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/inmunología , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/inmunología , Saccharomycetales/genética , Saccharomycetales/metabolismo , Anticuerpos de Dominio Único/biosíntesis , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/aislamiento & purificación , Anticuerpos de Dominio Único/farmacología
13.
Appl Environ Microbiol ; 86(8)2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32060024

RESUMEN

There is a growing need for a highly stable system to allow the production of biologics for diagnoses and therapeutic interventions on demand that could be used in extreme environments. Among the variety of biologics, nanobodies (Nbs) derived from single-chain variable antibody fragments from camelids have attracted great attention in recent years due to their small size and great stability with translational potentials in whole-body imaging and the development of new drugs. Intracellular expression using the bacterium Escherichia coli has been the predominant system to produce Nbs, and this requires lengthy steps for releasing intracellular proteins for purification as well as removal of endotoxins. Lyophilized, translationally competent cell extracts have also been explored as offering portability and long shelf life, but such extracts may be difficult to scale up and suffer from batch-to-batch variability. To address these problems, we present a new system to do the following: (i) engineer the spore-forming bacterium Bacillus subtilis to secrete Nbs that can target small molecules or protein antigens on mammalian cells, (ii) immobilize Nbs containing a cellulose-binding domain on a cellulose matrix for long-term storage and small-molecule capturing, (iii) directly use Nb-containing bacterial supernatant fluid to perform protein detection on cell surfaces, and (iv) convert engineered B. subtilis to spores that are resistant to most environmental extremes. In summary, our work may open a new paradigm for using B. subtilis as an extremely stable microbial factory to produce Nbs with applications in extreme environments on demand.IMPORTANCE It is highly desirable to produce biologics for diagnoses and therapeutic interventions on demand that could be used in a variety of settings. Among the many biologics, Nbs have attracted attention due to their small size, thermal stability, and broad utility in diagnoses, therapies, and fundamental research. Nbs originate from antibodies found in camelids, and >10 companies have invested in Nbs as potential drugs. Here, we present a system using cells of the bacterium Bacillus subtilis as a versatile platform for production of Nbs and then antigen detection via customized affinity columns. Importantly, B. subtilis carrying engineered genes for Nbs can form spores, which survive for years in a desiccated state. However, upon rehydration and exposure to nutrients, spores rapidly transition to growing cells which secrete encoded Nbs, thus allowing their manufacture and purification.


Asunto(s)
Bacillus subtilis/inmunología , Bioingeniería , Anticuerpos de Dominio Único/biosíntesis
14.
Biotechnol Bioeng ; 117(12): 3835-3848, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32808670

RESUMEN

Growth decoupling can be used to optimize the production of biochemicals and proteins in cell factories. Inhibition of excess biomass formation allows for carbon to be utilized efficiently for product formation instead of growth, resulting in increased product yields and titers. Here, we used CRISPR interference to increase the production of a single-domain antibody (sdAb) by inhibiting growth during production. First, we screened 21 sgRNA targets in the purine and pyrimidine biosynthesis pathways and found that the repression of 11 pathway genes led to the increased green fluorescent protein production and decreased growth. The sgRNA targets pyrF, pyrG, and cmk were selected and further used to improve the production of two versions of an expression-optimized sdAb. Proteomics analysis of the sdAb-producing pyrF, pyrG, and cmk growth decoupling strains showed significantly decreased RpoS levels and an increase of ribosome-associated proteins, indicating that the growth decoupling strains do not enter stationary phase and maintain their capacity for protein synthesis upon growth inhibition. Finally, sdAb production was scaled up to shake-flask fermentation where the product yield was improved 2.6-fold compared to the control strain with no sgRNA target sequence. An sdAb content of 14.6% was reached in the best-performing pyrG growth decoupling strain.


Asunto(s)
Sistemas CRISPR-Cas , Escherichia coli , Ingeniería Metabólica , Nucleótidos , Anticuerpos de Dominio Único/biosíntesis , Escherichia coli/genética , Escherichia coli/metabolismo , Nucleótidos/biosíntesis , Nucleótidos/genética , Anticuerpos de Dominio Único/genética
15.
Protein Expr Purif ; 172: 105645, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32289357

RESUMEN

Antibody fragments for which the sequence is available are suitable for straightforward engineering and expression in both eukaryotic and prokaryotic systems. When produced as fusions with convenient tags, they become reagents which pair their selective binding capacity to an orthogonal function. Several kinds of immunoreagents composed by nanobodies and either large proteins or short sequences have been designed for providing inexpensive ready-to-use biological tools. The possibility to choose among alternative expression strategies is critical because the fusion moieties might require specific conditions for correct folding or post-translational modifications. In the case of nanobody production, the trend is towards simpler but reliable (bacterial) methods that can substitute for more cumbersome processes requiring the use of eukaryotic systems. The use of these will not disappear, but will be restricted to those cases in which the final immunoconstructs must have features that cannot be obtained in prokaryotic cells. At the same time, bacterial expression has evolved from the conventional procedure which considered exclusively the nanobody and nanobody-fusion accumulation in the periplasm. Several reports show the advantage of cytoplasmic expression, surface-display and secretion for at least some applications. Finally, there is an increasing interest to use as a model the short nanobody sequence for the development of in silico methodologies aimed at optimizing the yields, stability and affinity of recombinant antibodies.


Asunto(s)
Expresión Génica , Pliegue de Proteína , Anticuerpos de Dominio Único , Animales , Humanos , Estabilidad Proteica , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Anticuerpos de Dominio Único/biosíntesis , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/aislamiento & purificación
16.
Appl Microbiol Biotechnol ; 104(11): 4837-4848, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32270250

RESUMEN

Variable domains of heavy chains of camelid heavy-chain antibodies (VHHs) are known as nanobodies. Nanobodies are approximately 15 kDa in size with high affinity to their antigens. They can be easily manipulated and produced in microorganisms. In this study, an alpaca was immunized with purified green fluorescence protein (GFP) and a VHH library from lymphocytes of the immunized alpaca was constructed with a capacity of 6.7 × 107. The library was biopanned against GFP by phage display technique and four unique DNA sequences coding for anti-GFP nanobodies were identified by enzyme-linked immunosorbent assay, named a12, e6, d5, and b9. The four DNA sequences were then cloned into pADL-10b-6×His or pBAD24-Flag-6×His for expression in bacteria. Purified A12, E6, D5, and B9 were demonstrated to bind GFP specifically both in vitro by enzyme-linked immunosorbent assay and native-PAGE analysis and in vivo by immunofluorescence and immunoprecipitation. Taken together, our results demonstrate that anti-GFP nanobodies are successfully selected from the immune library, are produced in bacteria, and are available for basic research.Key Points• Four different GFP binders were successfully obtained from an immune VHH library.• The four GFP binders were successfully purified from bacteria. • Purified GFP binders can bind GFP both in vitro and in vivo and are ready for use in basic research.


Asunto(s)
Camélidos del Nuevo Mundo/inmunología , Proteínas Fluorescentes Verdes/inmunología , Anticuerpos de Dominio Único/biosíntesis , Anticuerpos de Dominio Único/genética , Animales , Bacterias/genética , Sitios de Unión de Anticuerpos , Camelus , Técnicas de Visualización de Superficie Celular , Ensayo de Inmunoadsorción Enzimática , Biblioteca de Péptidos
17.
J Cell Biochem ; 120(6): 10787-10795, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30672018

RESUMEN

Adoptive transfer of T cells expressing chimeric antigen receptors (CARs) is considered to be a novel anticancer therapy. To date, in most cases, single-chain variable fragments (scFvs) of murine origin have been used in CARs. However, this structure has limitations relating to the potential immunogenicity of mouse antigens in humans and the relatively large size of scFvs. For the first time, we used camelid nanobody (VHH) to construct CAR T cells against prostate specific membrane antigen (PSMA). The nanobody against PSMA (NBP) was used to show the feasibility of CAR T cells against prostate cancer cells. T cells were transfected, and then the surface expression of the CAR T cells was confirmed. Then, the functions of VHH-CAR T cell were evaluated upon coculture with prostate cancer cells. At the end, the cytotoxicity potential of NBPII-CAR in T cells was approximated by determining the cell surface expression of CD107a after encountering PSMA. Our data show the specificity of VHH-CAR T cells against PSMA+ cells (LNCaP), not only by increasing the interleukin 2 (IL-2) cytokine (about 400 pg/mL), but also the expression of CD69 by almost 38%. In addition, VHH-CAR T cells were proliferated by nearly 60% when cocultured with LNCaP, as compared with PSMA negative prostate cancer cell (DU-145), which led to the upregulation of CD107a in T cells upto 31%. These results clearly show the possibility of using VHH-based CAR T cells for targeted immunotherapy, which may be developed to target virtually any tumor-associated antigen for adoptive T-cell immunotherapy of solid tumors.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Calicreínas/genética , Antígeno Prostático Específico/genética , Neoplasias de la Próstata/terapia , Receptores Quiméricos de Antígenos/genética , Anticuerpos de Dominio Único/química , Linfocitos T/inmunología , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos T/genética , Antígenos de Diferenciación de Linfocitos T/inmunología , Biomarcadores/metabolismo , Camelus , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Citotoxicidad Inmunológica , Electroporación , Expresión Génica , Humanos , Interleucina-2/genética , Interleucina-2/inmunología , Calicreínas/inmunología , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Proteína 1 de la Membrana Asociada a los Lisosomas/genética , Proteína 1 de la Membrana Asociada a los Lisosomas/inmunología , Masculino , Plásmidos/química , Plásmidos/inmunología , Cultivo Primario de Células , Próstata/inmunología , Próstata/patología , Antígeno Prostático Específico/inmunología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Receptores Quiméricos de Antígenos/inmunología , Anticuerpos de Dominio Único/biosíntesis , Anticuerpos de Dominio Único/aislamiento & purificación , Linfocitos T/citología
18.
Biochem Biophys Res Commun ; 519(2): 267-273, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31495493

RESUMEN

PD-1/PD-L1 pathway blocking with antibodies offers a vital and efficient therapeutic strategy to restore T cell-associated antitumor immunity and treats a variety of cancers in clinic. Nanobodies (Nbs) give several advantages over conventional monoclonal antibodies such as size, solubility, stability and costs. Additionally, P. pastoris is a suitable host for Nb production. Herein, we aim to produce and evaluate anti-PD-1 Nb derived from the P. pastoris. Our findings indicated that we successfully established the Nbs phage-displayed library against PD-1 with qualified library capacity and insert ratio. Anti-PD-1 Nb Nb97 was screened through PE-ELISA and flow cytometry. To extend half-life of Nb97, we contracted pPICZɑA-Nb97-Nb97-HSA recombination vector, which was then transformed into the system of P. pastoris X-33. The yield of purified Nb97-Nb97-Human serum albumin (HSA) fused protein (MY2935) reached to 2.3 g/L after 147 h of fermentation. Meanwhile, the blocking effect of MY2935 is similar to that of MY2626 (humanized Nb97-Fc), and MY2935 showed better performance on stimulating the immune function through PD-1 reporter assay. Hence, P. pastoris X-33 expressing and secreting functional anti-PD-1 Nb-HSA fusion protein might be a system of high yield and low cost.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Inmunoterapia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Saccharomyces cerevisiae/genética , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/inmunología , Células A549 , Antígeno B7-H1/inmunología , Línea Celular , Células HEK293 , Humanos , Receptor de Muerte Celular Programada 1/inmunología , Anticuerpos de Dominio Único/biosíntesis
19.
Appl Microbiol Biotechnol ; 102(2): 539-551, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29177623

RESUMEN

Camelid single-domain antibodies (sdAbs, VHHs, or Nanobodies®) are types of antibody fragments that are composed of the heavy-chain variable domain only. These VHHs possess unique structural and functional features, as they are small in size and exhibit thermal stability and high solubility. Compared to conventional antibodies, VHHs can be manufactured in microorganisms to significantly save on cost, labor, and time since VHHs lack the Fc domain with its N-linked oligosaccharide. Until now, VHHs have been expressed in several kinds of production systems, ranging from prokaryotic cells, yeasts, fungi, insect cells, and mammalian cell lines, to plants. In this review, we focus on the recent production of VHHs, introduce different platforms, and summarize the current state of this area and its future trends. Finally, the first potential VHH product, produced in Pichia pastoris, will probably be available on the market in 2018; thus, it is of great importance to give this antibody fragment timely attention. This is the first review concerning the production of VHHs in laboratory settings.


Asunto(s)
Ingeniería Genética , Anticuerpos de Dominio Único/biosíntesis , Animales , Línea Celular , Escherichia coli/metabolismo , Hongos/metabolismo , Cadenas Pesadas de Inmunoglobulina/biosíntesis , Pichia/metabolismo , Plantas/metabolismo
20.
Int J Mol Sci ; 19(11)2018 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-30400198

RESUMEN

Nanobodies, the smallest possible antibody format, have become of considerable interest for biotechnological and immunotherapeutic applications. They show excellent robustness, are non-immunogenic in humans, and can easily be engineered and produced in prokaryotic hosts. Traditionally, nanobodies are selected from camelid immune libraries involving the maintenance and treatment of animals. Recent advances have involved the generation of nanobodies from naïve or synthetic libraries. However, such approaches demand large library sizes and sophisticated selection procedures. Here, we propose an alternative, two-step approach for the design and generation of nanobodies. In a first step, complementarity-determining regions (CDRs) are grafted from conventional antibody formats onto nanobody frameworks, generating weak antigen binders. In a second step, the weak binders serve as templates to design focused synthetic phage libraries for affinity maturation. We validated this approach by grafting toxin- and hapten-specific CDRs onto frameworks derived from variable domains of camelid heavy-chain-only antibodies (VHH). We then affinity matured the hapten binder via panning of a synthetic phage library. We suggest that this strategy can complement existing immune, naïve, and synthetic library based methods, requiring neither animal experiments, nor large libraries, nor sophisticated selection protocols.


Asunto(s)
Ingeniería de Proteínas/métodos , Anticuerpos de Dominio Único/biosíntesis , Anticuerpos de Dominio Único/química , Secuencia de Aminoácidos , Animales , Antígenos/metabolismo , Camelus , Regiones Determinantes de Complementariedad , Fluoresceína/metabolismo , Haptenos/metabolismo , Cadenas Pesadas de Inmunoglobulina/química , Cadenas Pesadas de Inmunoglobulina/metabolismo , Región Variable de Inmunoglobulina/química , Biblioteca de Péptidos , Toxinas Biológicas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA